Wednesday 08 October |
08:30 |
"Wednesday 08 October"
Added to your list of favorites
Deleted from your list of favorites
A01
08:30 - 17:00
PRE CONGRESS WORKSHOP 1
Preclinical imaging
|
Espace Vieux-Port |
|
"Wednesday 08 October"
Added to your list of favorites
Deleted from your list of favorites
B01
08:30 - 17:00
PRE CONGRESS WORKSHOP 2
Microstructural imaging
09:00 - 09:15
OPENING.
09:15 - 09:40
Diffusion Microstructure.
Andrada IANUS (Keynote Speaker, United Kingdom)
09:40 - 10:05
Principles of Relaxation and QSM.
Valerij KISELEV
10:05 - 10:30
Dealing with Motion in Body Diffusion Imaging.
Dimitrios KARAMPINOS
10:30 - 10:45
BREAK.
10:45 - 11:10
Diffusion NMR in Porous Media: Advances and Perspectives.
Denis GREBENKOV (Keynote Speaker, France)
11:10 - 11:35
Virtual Tissues, Real Insights: Simulating Biological Microstructure for MRI.
Mariam ANDERSSON
11:35 - 12:00
Simulating Diffusion and Perfusion in Microvasculature.
Elizabeth POWELL
12:00 - 12:20
A round table discussion from three different points of view.
Benedicte MARECHAL (Research scientist) (Keynote Speaker, Lausanne, Switzerland)
12:20 - 13:30
Corporate symposium: Advanced Diffusion and Tractography in Olea Sphere: A Gateway to Microstructural Imaging.
Stefano CASAGRANDA
13:30 - 14:45
Data handling, pre-processing and pipelining.
Oscar ESTEBAN
14:45 - 15:30
Break and POSTER SESSION #1.
15:30 - 15:55
Microstructural diffusion MRI as a functional contrast complementary to BOLD.
Ileana JELESCU
15:55 - 16:20
Seismology of the Brain.
Jaco JM ZWANENBURG
16:20 - 16:45
Emerging AI Methods for Parameter Estimation.
Maeliss JALLAIS (Research Associate) (Keynote Speaker, Cardiff, United Kingdom)
16:45 - 16:55
Concluding remarks.
17:00 - 17:00
POSTER SESSION #2.
|
Auditorium 900 |
|
"Wednesday 08 October"
Added to your list of favorites
Deleted from your list of favorites
C01
08:30 - 17:00
PRE CONGRESS WORKSHOP 3
GREC (Gadolinium Research & Education Committee)
|
Salle Major |
|
"Wednesday 08 October"
Added to your list of favorites
Deleted from your list of favorites
E01
08:30 - 17:00
PRE CONGRESS WORKSHOP 5
CAMERA Workshop
|
Salle 50 |
|
"Wednesday 08 October"
Added to your list of favorites
Deleted from your list of favorites
F01
08:30 - 17:00
PRE CONGRESS WORKSHOP 6
MR Safety
|
Salle 76 |
09:00 |
"Wednesday 08 October"
Added to your list of favorites
Deleted from your list of favorites
D01
09:00 - 17:00
PRE CONGRESS WORKSHOP 4
GliMR (Glioma MR)
The first 25 junior registrants under 35 will receive reimbursement for the
precongress and ESMRMB main congress worth 130 Euros!
Register for GliMR via this link: https://forms.gle/sGXGjAgqtfoRg7tp7
(note if you are a junior while filling in)
Pay for the First GliMR Congress via the ESMRMB 2025 registration portal from MCO: https://esmrmb2025.mycongressonline.net/13-Home_reg.html
09:00 - 09:05
Casual walk-in.
09:05 - 09:10
Opening welcome.
Esther WARNERT (Chairperson, The Netherlands)
09:10 - 09:40
Power Pitches of GliMR Task Force leads presenting their activities.
The Task Force Leads
09:40 - 10:30
Key topic 1: Tumoral microenvironments: what to see and how to interpret (Use case: tumor progression).
Rui SIMÕES (Keynote Speaker, Porto, Portugal), Tom BOOTH (Keynote Speaker, London, United Kingdom), Harish POPTANI (Keynote Speaker, Liverpool, United Kingdom)
10:30 - 11:05
Coffee Break.
11:05 - 12:35
Hands-on workshop on individualised perfusion imaging in brain tumors.
12:35 - 13:30
Lunch break.
13:30 - 14:00
Keynote lecture: "Imaging follow-up in IDH-mutated glioma patients: from tumor kinetics to brain plasticity".
Emmanuel MANDONNET (Keynote Speaker, France)
14:00 - 14:45
Key Topic 2: What is going on here? My weirdest case.
Gilbert HANGEL (Keynote Speaker, Australia), Patricia CLEMENT (PhD) (Keynote Speaker, Ghent, Belgium), Rita G. NUNES (PhD) (Keynote Speaker, Lisbon, Portugal), Albert PONS-ESCODA (Senior Consultant Neuroradiologist) (Keynote Speaker, Barcelona, Spain)
In this format, a panel of experts and the audience will try to solve anomalies in cases, data, statistics
and images. Having mystery cases or data? Bring it in to this clinic! Send your case or data issue to:
v.c.w.keil@amsterdamumc.nl
14:45 - 15:30
Key topic 3: IDH mutation prediction - Where are we now?
Johannes SLOTBOOM (Keynote Speaker, Switzerland), Esin Öztürk ISIK (Keynote Speaker, Turkey)
The clinical and the technological perspective with group discussion
15:30 - 16:00
Coffee Break.
16:00 - 16:45
Break-out Sessions of the Task forces and poster walk for project abstracts.
Get to know the GliMR task forces and their projects for European glioma imaging better. Get involved - or present a poster with an intended project at the project poster wall.
TF 1: Advanced Imaging | TF 2: Preclinical Imaging |TF 3: AI | TF5: Clinical Implementation | TF 8: Science Communication
16:45 - 17:00
Closing remarks.
Esther WARNERT (Chairperson, The Netherlands)
|
Salle 120 |
Thursday 09 October |
09:00 |
"Thursday 09 October"
Added to your list of favorites
Deleted from your list of favorites
A10
09:00 - 09:30
OPENING
|
Auditorium 900 |
09:30 |
"Thursday 09 October"
Added to your list of favorites
Deleted from your list of favorites
A11
09:30 - 10:30
FT3 Plenary
Good for what? Defining quality from a practical perspective
FT1: Cycle of Technology
09:30 - 10:30
Quality control of AI methods: Evidence and evaluation.
Gaël VAROQUAUX (Keynote Speaker, France)
09:30 - 10:30
Towards high quality MR biomarkers.
Martina CALLAGHAN (PhD) (Keynote Speaker, London, United Kingdom)
|
Auditorium 900 |
10:30 |
"Thursday 09 October"
Added to your list of favorites
Deleted from your list of favorites
E11
10:30 - 13:30
GREC
|
Salle 76 |
|
TIME FOR A BREAK
- Coffee and refreshments will be available at the cash bar.
|
11:00 |
"Thursday 09 October"
Added to your list of favorites
Deleted from your list of favorites
A13
11:00 - 12:00
FT1 ORAL - Technology to study brain dynamics
FT1: Cycle of Technology
11:00 - 11:10
#46421 - PG001 Exploring functional connectivity and resting-state networks within isotropic ADC-fMRI.
PG001 Exploring functional connectivity and resting-state networks within isotropic ADC-fMRI.
Resting-state (RS) functional MRI (fMRI) [1] captures coherent spatial patterns called Resting State Networks (RSNs) [2], characterised by synchronous Blood Oxygen Level-Dependent (BOLD) fluctuations [3]. Functional connectivity (FC) analysis typically shows high correlations amongst grey matter (GM) regions within the same RSN. Some studies have extended FC analysis by including white matter (WM) regions [4-6]. However, the incorporation of WM analysis with BOLD fMRI is challenging due to the more limited vasculature and altered hemodynamic response function (HRF) in WM [5].
To overcome BOLD-fMRI limitations, recent task-based studies explored Apparent Diffusion Coefficient (ADC)-fMRI, which is independent of vascular response and equally sensitive to neural activity in both GM and WM [7–9]. Earlier RS analysis [10] demonstrated BOLD-like FC in GM and high sensitivity to WM connections for ADC-fMRI, but was limited by linear diffusion encoding and partial brain coverage.
We aim to study FC using full brain coverage and diffusion spherical tensor encoding [11] for ADC-fMRI, allowing isotropic sensitivity to neural activity. We extract ADC RSNs via Independent Component Analysis (ICA) and compare them with BOLD RSNs.
RS BOLD and ADC-fMRI were acquired from 13 subjects with parameters summarised in Table 1. BOLD and dMRI data were preprocessed as described in [10]. ICA cleaning (FSL MELODIC) and motion correction were applied separately to BOLD, dMRI b = 200 and b = 1000 time series. ADC-fMRI time series were computed from dMRI as in [8-11], and the data proven to be largely free of vascular contamination [12].
FC matrices were computed using Pearson correlations (PC) between GM (Desikan-Killiany) and WM (Juelich) ROIs, including only positive connections. After Fisher transformation, a one-sample t-test (Benjamini-Hochberg corrected, α = 0.05) identified significant group-level connections, and a threshold at mean + 2.3 × SD was computed. Subject-specific FC matrices were binarised at this threshold for subsequent graph analysis using Networkx. Inter-subject consistency was measured as the mean PC across pairs of unthresholded individual FC matrices.
Finally, group ICA was performed to extract RSNs, using 100 components for BOLD [13], while FSL automatically determined 250 components for ADC. Figure 1 shows the averaged FC matrices for BOLD and ADC. BOLD exhibits stronger overall correlations with prominent GM-GM connectivity, particularly across hemispheres. ADC FC shows weaker but comparable GM-GM connectivity, especially within the DMN and motor networks, and features more relatively important WM-GM connections than BOLD. In Figure 2, the average clustering coefficient is significantly higher in BOLD than in ADC for GM-GM and WM-GM. For global efficiency and average node degree, ADC has higher values than BOLD, with significance only occurring for the global efficiency in WM-GM. Graph metric variability is generally lower in ADC-fMRI than in BOLD, suggesting greater inter-subject consistency. WM-GM FC shows higher between-subject similarity in ADC (PC = 0.37 for ADC vs. 0.44 for BOLD, p=0.003), whereas BOLD is more consistent for WM-WM (PC = 0.56 vs. 0.40, Wilcoxon p=10⁻⁹) and GM-GM (PC = 0.60 vs. 0.58, p=0.12) FC. The main RSNs identified by ICA with BOLD can also be identified with ADC fMRI, though some RSNs that appear as single components in BOLD are split across multiple components in ADC (Figure 3). Although ADC-fMRI is independent of vascular contributions, FC analysis shows that it captures positive correlations comparable to BOLD-fMRI, supporting its utility for resting-state studies. Although BOLD shows overall stronger correlations, the strongest connections are largely consistent across both modalities. In particular, ADC demonstrates sensitivity to expected connections within the DMN. Amongst the strongest connections, WM-GM links appear more frequently in ADC than in BOLD, possibly due to the delayed BOLD HRFs in deep WM [14].
In line with previous findings [10], ADC-fMRI demonstrates higher average node degree and global efficiency in all types of connections. Although higher between-subject similarity in ADC than in BOLD for WM-WM connectivity was reported [10], our results show this pattern instead for WM-GM connections.
The overlap between ADC and BOLD ICA components confirms that ADC-fMRI captures meaningful RSNs. Fragmented clusters in ADC may stem from lower SNR, though FC remained stable due to region-level averaging. FC analysis shows similarities between BOLD and ADC, notably the strong connections within the DMN. Our results also suggest that ADC is more sensitive to GM-WM connectivity than BOLD, as these connections are more frequently among the strongest and show greater inter-subject consistency. Although RSNs appear fragmented across ADC independent components, this first demonstration encourages further investigation such as seed-based analysis and co-activation patterns.
Jasmine NGUYEN-DUC (Lausanne, Switzerland), Arthur SPENCER, Jean-Baptiste PEROT, Inès DE RIEDMATTEN, Filip SZCZEPANKIEWICZ
11:10 - 11:20
#47903 - PG002 Decomposing Brain State Dynamics: A Spectral and Temporal Characterization of Resting fMRI.
PG002 Decomposing Brain State Dynamics: A Spectral and Temporal Characterization of Resting fMRI.
Understanding the brain’s intrinsic functional architecture requires methods that capture both its temporal and spectral properties. However, conventional resting-state fMRI analyses often rely on fixed frequency bands or static models, limiting insight into how intrinsic oscillatory timescales influence functional brain states [1-2]. To address this, we employ Empirical Mode Decomposition (EMD) [3], a data-driven approach that decomposes BOLD signals into intrinsic mode functions (IMFs) without assuming predefined spectral boundaries, enabling frequency-resolved analysis of dynamic functional connectivity. Using instantaneous phase estimation [4] and clustering of leading eigenvectors from phase coherence matrices [5], it is possible to identify recurrent brain states across oscillatory scales. Prior work by Goldhacker et al. demonstrated that functional brain states can remain stable across frequencies, highlighting the multiscale nature of brain networks [6]. This study aims to characterize how brain state dynamics vary across intrinsic frequency scales.
We analyzed resting-state fMRI data from 96 healthy adults from the Human Connectome Project [7] to investigate the spectro-temporal organization of intrinsic brain states. The fMRI data were parcellated into 116 regions (100 cortical and 16 subcortical) [8-9] To extract oscillatory components, we applied EMD to the BOLD time series, yielding IMFs that represent frequency-specific fluctuations [3]. For each IMF and parcel, instantaneous phase time series were computed using the Hilbert transform [4]. Phase coherence matrices were then constructed at each time point, and their leading eigenvectors were extracted using singular value decomposition [5], capturing dominant phase alignment patterns across the brain. These eigenvectors were clustered using k-means to identify recurrent brain states. We then computed dynamic metrics for each state, including the probability of occurrence, mean lifetime, and transition probabilities [5]. To assess the frequency-dependent modulation of brain state dynamics, we conducted generalized linear mixed-effects models (GLME) comparing the probability of occurrence and mean lifetime of each state across five IMFs. We extracted five IMFs with peak frequencies centered at ~0.01, 0.02, 0.04, 0.08, and 0.18 Hz (IMF5 to IMF1, Fig.1). Clustering the leading eigenvectors of phase coherence matrices revealed four recurrent brain states, with the optimal number determined via silhouette analysis (Fig.2). These states mostly mapped onto distinct functional systems: (1) visual network, (2) somatomotor network, (3) attention networks (dorsal and salience/ventral), and (4) a default mode and executive control network (DMN/ECN). The GLME results revealed significant main effects of IMF on both the probability of occurrence and lifetime for all four states (p < 0.001). Post-hoc comparisons showed that the visual and somatomotor states exhibited a higher probability of occurrence at higher-frequency IMFs, while the attentional and DMN/ECN states became more prominent at lower-frequency IMFs. Overall, the mean lifetime of all states decreased with increasing frequency, indicating reduced temporal stability at faster timescales (Fig.3). Moreover, state transition matrices revealed frequency-dependent differences in switching behavior (Fig.4). Lower-frequency IMFs exhibited fewer transitions, indicating greater metastability. In contrast, higher-frequency IMFs showed more frequent transitions between states, reflecting greater dynamical flexibility. Our findings reveal that the brain's dynamic functional architecture is hierarchically organized across frequency scales. Higher-order brain states, such as those involving attentional and DMN/ECN were more dominant and stable at slower frequencies. In contrast, unimodal states, particularly those associated with visual and somatomotor networks, emerged more prominently at faster frequencies. These findings support the notion that distinct functional systems operate on characteristic timescales, likely reflecting their respective roles in cognitive integration versus sensory processing. Furthermore, we observed greater metastability in lower-frequency IMFs, characterized by fewer transitions and longer lifetimes within each state. Conversely, higher-frequency IMFs were associated with more frequent switching between states, indicating enhanced dynamical flexibility. This frequency-dependent organization aligns with theoretical models proposing that the brain balances integration and segregation through activity at different temporal scales. This frequency-resolved approach provides new insights into the temporal structure of functional brain states, highlighting the importance of spectral decomposition in understanding the organization of dynamic connectivity. Future work may explore the behavioral or clinical relevance of these spectro-temporal profiles across populations and cognitive phenotypes.
Hüden NEŞE (Üsküdar, Turkey), Ahmet ADEMOĞLU, Tamer DEMIRALP
11:20 - 11:30
#48041 - PG003 Task-Evoked Dynamics and EEG–fMRI Coupling Under Autonomic Nuisance Regression.
PG003 Task-Evoked Dynamics and EEG–fMRI Coupling Under Autonomic Nuisance Regression.
Functional MRI (fMRI) BOLD signals are known to correlate with autonomic measures, including respiratory volume, heart rate, and vascular tone (Birn et al.,2006, Chang et al.,2009, Özbay et al.,2018). While these signals are often treated as non-neuronal confounds, emerging evidence suggests they may instead reflect a shared arousal-related process that influences both cortical and autonomic activity (Gu et al.,2022). Although such autonomic contributions have been well characterized during rest and sleep, their role in cognitive task performance remains underexplored.
Multimodal data were collected (N=5) during a block-design mental arithmetic task (solving an equation with one unknown), including 3T fMRI (TR=3s, TE=36ms, FA=90, spatial resolution=2.5mm, scan time=135TR), EEG, respiratory, and photoplethysmography (PPG) signals. Autonomic regressors—respiratory volume per time (RVT) and PPG amplitude (ppgA)—were extracted from the physiological recordings and their time-shifted versions were used in nuisance regression (autonomic correction as in Picchioni et al., 2022) alongside motion parameters and RETROICOR regressors (Glover et al., 2000) (Figure 1). Residual BOLD signals were subjected to event-locked averaging and cross-modal analyses. Event-related group-level z-maps were computed at representative timepoints post-task onset under two preprocessing models: RETROICOR only and RETROICOR + RVT + ppgA (Figure 2). Block-averaged time courses of BOLD, RVT, and ppgA signals were also examined. To assess EEG–fMRI coupling, voxelwise cross-correlation analyses were performed between z-scored EEG spectral power (e.g., alpha, beta bands) and preprocessed BOLD signals across preprocessing conditions. Subject-level cross-correlation maps were generated (Figure 3) and averaged after Fisher Z-transformation for group-level analysis (Figure 4). Representative results are shown to illustrate task-correlated changes in all modalities and the effect of autonomic correction on EEG-fMRI coupling. Event-locked 3D maps showed BOLD increases around 6 seconds after task onset—consistent with the hemodynamic response—in regions including the IPS, visual cortex, cuneus, insula, thalamus, MCC, and posterior cingulate, followed by a decrease around 15 seconds in the precuneus. These task-related BOLD changes were attenuated after correction with RVT and PPG amplitude. Group-level block-averaged time courses showed that the BOLD increase was accompanied by a sharp drop in PPG amplitude, indicating sympathetic vasoconstriction, while no consistent task-related change in RVT was observed.
Cross-correlation analyses between EEG spectral power and BOLD at +9 s lag (EEG leads) revealed negative correlations in regions such as the IPS and visual cortex at the subject level, which diminished after autonomic correction, particularly in individuals with marked task-related autonomic responses. At the group level, negative correlations between frontal-occipital beta and occipital alpha power and BOLD in regions including the insula, thalamus, retrosplenial cortex, and periventricular areas were reduced following correction with RVT and PPG amplitude. Additionally, block-averaged EEG alpha and beta power showed slight task-related changes at the group-level suggesting arousal modulation post-task onset. Our results show that autonomic fluctuations, particularly those linked to sympathetic arousal (e.g., PPG amplitude), contribute to both task-evoked BOLD changes and EEG–fMRI coupling. Event-locked and cross-modal analyses revealed that these task-related BOLD responses and late EEG–BOLD correlations are reduced after correcting for RVT and PPG amplitude. Importantly, many of the affected brain regions, such as the thalamus, insula, and cingulate cortex, are key components of the central autonomic network (Beissner et al.,2013), suggesting a shared functional pathway linking autonomic regulation and neural activity. The partial reduction of late negative EEG–BOLD correlations following correction suggests these relationships are not purely neural but may reflect integrated arousal mechanisms modulating both systemic and neurogenic activity. Inter-subject variability further underscores individual differences in the expression and impact of these processes. These findings underscore the importance of modeling autonomic influences when interpreting fMRI and EEG–fMRI data, not only in resting-state or sleep studies but also during cognitively demanding tasks. Autonomic and arousal-related fluctuations can confound neural interpretations of BOLD activity and contribute to inter-subject variability. Future studies should aim to disentangle neurovascular from systemic vascular contributions to improve the precision of multimodal brain imaging analyses.
Kübra EREN, Lina ALQAM, Belal TAVASHI, Kadir Berat YILDIRIM, Elif CAN, Cem KARAKUZU, Alp DINÇER, Pınar ÖZBAY (İstanbul, Turkey)
11:30 - 11:40
#46101 - PG004 Tract-specific diffusion MRI correlates of electrophysiological sensory impairments in asymptomatic cervical cord compression subjects: preliminary study.
PG004 Tract-specific diffusion MRI correlates of electrophysiological sensory impairments in asymptomatic cervical cord compression subjects: preliminary study.
Asymptomatic degenerative cervical cord compression (ADCC) represents a premyelopathic stage of degenerative cervical myelopathy (DCM), highly prevalent in the aging population and lacking clear clinical management guidelines [1]. ADCC affects up to 40% of individuals [2], with approximately one-quarter progressing to symptomatic DCM within 44‐months [3, 4]. Early detection of subclinical changes in spinal cord function may be critical to identify individuals at risk of progression. Diffusion MRI (dMRI) enables tract-specific assessment of spinal cord microstructure, offering potential biomarkers of early changes [5–8]. Electrophysiological techniques such as somatosensory evoked potentials (SEPs) and contact heat evoked potentials (CHEPs) provide sensitive measures of dorsal column and spinothalamic tract dysfunction, respectively [9, 10]. This study investigates the utility of tract-specific dMRI in ADCC subjects exhibiting electrophysiological abnormalities in the spinal cord sensory system.
This retrospective study included 74 ADCC subjects recruited at the University Hospital Brno, Czechia, who underwent clinical, MRI, and electrophysiological examinations. MRI was acquired on a 3T Siemens Prisma scanner. For the purpose of this study, we used T2-weighted (T2-w) 0.8 mm isotropic scans [11] covering the whole cervical spine and multi-shell dMRI scans covering C3-C6 vertebral levels [12]. Spinal cord compression was visually assessed by two experienced radiologists. In cases of multilevel compression, the maximum compression level (MCL) was defined as the level with the lowest compression ratio [13].
Electrophysiological examinations included short-latency SEPs (median and tibial nerves, bilaterally) and bilateral dermatomal CHEPs (C4, C6 C8) [3, 14–16]. SEPs abnormality was defined as the presence of any abnormality. CHEPs abnormality was defined as abnormality of N2 latency and/or N2/P2 amplitude or absent N2/P2 waveform.
Figure 1 shows automatic MRI data processing using Spinal Cord Toolbox (SCT) [17] v7.0. For T2-w images, the spinal cord was segmented [18], intervertebral discs were identified [19], and registration to the PAM50 template [20] was performed. dMRI images were motion corrected and denoised [21], the spinal cord was segmented [18], and the PAM50 template and white matter atlas were registered to the dMRI space using the T2w-to-template transformation as initial transformation. All processing steps were visually verified and corrected if necessary. Diffusion tensor imaging (DTI) metrics, including fractional anisotropy (FA), mean, (MD), axial (AD), and radial diffusivities (RD), were computed and extracted within dorsal columns and spinothalamic tracts between C3 and C6. DTI metrics from the dorsal columns were compared between subjects with normal and abnormal SEPs. Similarly, DTI metrics from the spinothalamic tracts were compared between subjects with normal and abnormal CHEPs. The demographic, clinical, imaging and electrophysiological characteristics are summarized in Table 1. Comparison of DTI metrics extracted from the dorsal columns revealed significantly (p<.05) lower FA and higher RD in subjects with abnormal SEPs compared to those with normal SEPs. Additionally, subjects with abnormal SEPs showed non-significant trends toward higher AD and MD (Figure 2). No significant differences were observed in DTI metrics within the spinothalamic tracts between subjects with normal and impaired CHEPs (data not shown). Decreased FA and increased RD were previously reported in subjects with compression compared to healthy controls [5, 22–24]. In our study, we observed similar changes in the dorsal columns of ADCC subjects with abnormal SEPs. As SEPs abnormalities were caused predominantly due to prolonged latencies, which may be associated with myelin loss [25], and decreased FA along with increased MD and RD have been linked to demyelination [24], we hypothesize that the DTI changes observed in the dorsal columns may reflect early demyelination of these long, highly myelinated tracts.
In contrast, we did not find any significant changes in DTI metrics between subjects with normal and impaired CHEPs. This may be due to the smaller, less-myelinated axons of the spinothalamic tracts, making them less susceptible to demyelination. Notably, the clinical-radiological paradox, where individuals show MRI evidence of cord compression without clinical symptoms, has also been documented [26, 27].
Future work will aim for more granular, laterality-specific analyses and increased sample sizes, to enable group matching and reduce selection bias. High-resolution 3T dMRI enables tract-specific analysis of spinal cord microstructure in relation to electrophysiological assessments. The findings for the dorsal columns are consistent with previous literature; still, further research is needed to gain more insights into the relationship between MRI and electrophysiological measures.
Jan VALOSEK (Montreal, Czech Republic), Eva VLČKOVÁ, Miloš KEŘKOVSKÝ, Tomáš ROHAN, Julien COHEN-ADAD, Josef BEDNAŘÍK
11:40 - 11:50
#47648 - PG005 Cerebrospinal fluid fluctuations from vasomotion in relation to cortical thinning in patients with mild cognitive impairment and dementia.
PG005 Cerebrospinal fluid fluctuations from vasomotion in relation to cortical thinning in patients with mild cognitive impairment and dementia.
Dementia poses an increasing global healthcare burden. Dementias with underlying neurovascular(1) or neurodegenerative disease (e.g. Alzheimer’s disease (AD))(2) are partly characterized by metabolic waste protein accumulation in brain tissue. Impaired clearance of brain waste is therefore related to these diseases(3). In neurovascular or neurodegenerative diseases, vasomotion as a driver of cerebrospinal fluid (CSF) into brain tissue might be impaired(3–5). This impairment could be related to two other MRI markers of these diseases(6), namely cortical thinning and white matter hyperintensities (WMH). We therefore aimed to study the association between cortical thinning and low-frequency vasomotion-related BOLD fMRI (0.1 Hz(7) or lower(8)) as well as CSF signals gained from inflow-sensitized fast fMRI signal in the fourth ventricle(9). We also compared BOLD-CSF coupling(9) between patients with mild cognitive impairment (MCI) and dementia. Lastly, we related WMH burden to CSF signal fluctuations.
Memory clinic patients (n=16, mean age 74.5±4.5 SD) with a clinical diagnosis of MCI (n=8) or dementia (n=8) were prospectively recruited at the Leiden University Medical Center (LUMC), the Alrijne hospital, or the Haga hospital, in the Netherlands. Patients were scanned at 3T and 7T MRI on the same day at the Leiden University Medical Center. For all scan parameters, see Table 1.
CSF signal was extracted by manually drawing on the standard deviation of the bottom slice(9). For details on post-processing see Figure 1. The power spectral density (PSD) was calculated for BOLD and CSF signal and the area under the curve (AUC) of the low-frequency range was taken (<0.125 Hz). These were Spearman correlated to cortical thickness. For BOLD-CSF coupling, BOLD and CSF signals were cross-correlated. We found no significant Spearman correlation between cortical thickness and the AUC PSD of the BOLD signals nor CSF signals (all p>.05), not for the total cerebral cortex nor for secondary analyses in temporal, occipital, parietal, and frontal lobes (Figure 2). Mean cortical thickness was 1.9±0.2mm.
Dementia and MCI patient groups showed comparable age, BMI and mean arterial pressure (rank sum test, all p>.05). Patients with dementia had generally lower memory test scores and MMSE scores.
BOLD-CSF correlation curves were calculated for dementia and MCI groups (Figure 3). Peak correlation magnitude was slightly higher for the dementia group (mean diff.=0.11, rank sum (Mann-Whitney U) test p=.049). Lags did not statistically differ between patient groups (rank sum test p>.05).
Mean WMH volume was 19±21ml (SD). WMH volume was not associated with the AUC of low-frequency CSF fluctuations (rank sum test p>.05). Dementia and MCI groups did not have significantly different WMH volumes (rank sum test p>.05) In a small prospective sample of MCI and dementia patients, we could not establish a relation between cortical thinning and signal proxies of brain vasomotion and (global) CSF fluctuations, respectively. This tentatively suggests that vasomotion and CSF fluctuations are not directly affected by cortical thinning. However, these results should be confirmed in studies with a larger sample size.
We also found that the BOLD-CSF peak correlation magnitudes were higher for the dementia group than the MCI group. This suggests that the cortical BOLD and CSF signals of dementia patients are more in synchrony. Dementia patients were initially expected to have lower BOLD-CSF coupling than MCI patients(5) possibly due to vasomotor dysfunction. However, neurovascular risk factors such as WMH volumes, age, BMI, and MAP or hypertensive status were similar. Perhaps this result is a false positive as a limitation of the small sample.
Sequence choices can influence flow measurements. Thick axial slices (5.5 mm) accelerate signal sampling but can also reduce CSF signal amplitudes compared to thinner slices(10,11). Thick slices also give more partial voluming. Still, BOLD-CSF correlations were higher than previous averages of AD and MCI(5), likely due to faster fMRI.
Low-frequency CSF fluctuations were not associated with mean WMH volume. While WMH-volume is a robust structural marker of small vessel disease, it is possible that this relationship cannot capture all variability related to functional BOLD signals.
In general, more prospective data collection is needed to substantiate these findings. In a small prospective cohort study of patients with dementia and MCI, we found no relation between cortical thinning and fast fMRI signals approximating cortical vasomotion and CSF fluctuations. BOLD and CSF signals were more in synchrony for patients with dementia than with MCI. CSF signals seem to be unrelated to white matter hyperintensity burden.
Ingmar EILING (Leiden, The Netherlands), Jasmin KUHN-KELLER, Lydiane HIRSCHLER, Emiel ROEFS, Marie-Noëlle WITJES-ANÉ, Marjan VAN DER ELST, Evelien SOHL, Joep LAGRO, Simon MOOIJAART, Jeroen DE BRESSER, WHIMAS STUDY GROUP
11:50 - 12:00
#47626 - PG006 Can real-time phase contrast quantification of CSF flow predict shunt response in NPH patients?
PG006 Can real-time phase contrast quantification of CSF flow predict shunt response in NPH patients?
The prevalence of idiopathic normal-pressure hydrocephalus (iNPH) is 1.5% for 70-year-olds, and increases with age [1]. The symptoms of gait disturbances, urinary incontinence and cognitive decline can often be reversed by surgically placing a shunt that removes cerebrospinal fluid (CSF) from the ventricular system. However, prediction of shunt response is poor and studies investigating the use of cardiac-gated phase contrast (CG PC) MR for outcome prediction have been inconclusive [2-4]. However, it is shown that iNPH patients have wider cerebral aqueducts, faster CSF flow and larger net flow volumes than age matched controls, and that these parameters can be restored to normal values by interventions such as lumbar tap test and shunt placement [5-8].
It is shown that respiration has a large influence on CSF flow [9-12]. In contrast to CG PC, real-time phase-contrast (RT PC) preserves the respiratory effects on CSF flow dynamics. RT PC methods are mainly validated in healthy volunteers, and their utility in clinical settings is unknown.
This study investigates if CSF flow measured with RT PC prior to shunt placement correlates with clinical outcome of NPH-patients.
iNPH patients (N=19) scheduled for shunting underwent a series of clinical tests and a 7T MR examination (Philips 7T Achieva). The study was approved by the Swedish Ethical Review Authority and all subjects gave written informed consent. Cerebral aqueduct flow was measured with RT PC using a golden-angle radial trajectory [13]. In addition, a CG PC scan was acquired (Fig. 1). RT data was reconstructed offline using the MRecon (Gyrotools) and BART [refs]. Data analysis was performed in Segment (v4.1 R14708 Medviso [14]) with custom plugins for real-time analysis. Data was corrected for linear phase background and unaliased in cases of phase wraps. RT flow curves were low-pass filtered to remove the influence of cardiac pulsation and forward, backward and net flows were extracted (Fig. 2). Net flow was computed as the difference between antegrade and retrograde flow volumes, and stroke volume (SV) was defined as the sum of the flow volumes divided by 2.
Clinical outcome was measured as percentage changes in test scores of motor and cognitive functions before (64±23 weeks) and after (16±3 weeks) intervention (Fig. 3). A normative sample was created using a bootstrap procedure, and the patients' individual changes were then relatively ranked based on the performance of that sample. For each clinical test, percentage change was and classified as '' Improved'', ''Worsened'' or ''Inconclusive'' based on a threshold of 95% CI in the normative sample (Fig. 3C). An improvement score was derived by subtracting the number of ''Worsened'' tests from the ''Improved'' ones and normalising by the total number of tests. A positive score indicates improved performance on the tests. A statistically significant correlation was found between real-time CSF net flow and normalised improvement score (p-value = 0.028, r=0.50), with craniocaudal (antegrade) flow associated with favourable outcomes. In contrast, no significant correlations were found between RT CSF volume, CG net flow or CG SV and normalised improvement score (Fig. 4). A correlation analysis between percentage changes of the individual tests and RT net flow gave no statistically significant results, except for MMSE-SR (p-value= 0.05). The use of 7T MR allows for high spatial resolution in both CG and RT PC, which is required to avoid that partial volume effects skew the results [15]. For radial RT PC, the desired temporal resolution is selected in the image reconstruction and is here set to be sufficient to investigate the slowly varying CSF flow. However, the study is limited by a small and heterogenous cohort, and recruitment of more patients as well as age matched controls is ongoing. In future analysis, other variables such as velocity and pulsatility will be investigated. Breathing has a large influence on the CSF flow, and this study was performed in free breathing. Performing the RT PC under guided breathing could reduce inter- and intraindividual variations in breathing patterns. A recent study investigates CSF flow in several anatomical locations iNPH patients using an EPI-based RT PC method, showing differences in patients and controls [16]. Interestingly, they conclude that disordered breathing may be a contributing factor in iNPH pathophysiology, supporting the use of RT PC rather than CG PC in this patient group. Our results suggest that real-time PC CSF net flow measurements in the aqueduct are indicative of the outcome of shunt placement in iNPH patients, with antegrade net flow being correlated to clinical improvement. A larger study could confirm the result and determine the clinical relevance of the method.
Federica CALAFIORE, Sara HALL, Mattis JALAKAS, Nicola SPOTORNO, Danielle VAN WESTEN, Niklas MARKLUND, Johannes TÖGER, Karin MARKENROTH BLOCH (Lund, Sweden)
|
Auditorium 900 |
|
"Thursday 09 October"
Added to your list of favorites
Deleted from your list of favorites
B10
11:00 - 12:00
FT2-4 ADVANCED MR SPECTROSCOPY
FT2: Cycle of Translation
11:00 - 12:00
In-vivo 2D NMR.
Lucio FRYDMAN (Keynote Speaker, Israel)
11:00 - 12:00
#45841 - NMR spectroscopy in biomedical applications – from lab to clinics.
NMR spectroscopy in biomedical applications – from lab to clinics.
Nuclear Magnetic Resonance (NMR) spectroscopy has become an essential tool in studying human metabolism, providing deep insights into the biochemical foundations of health and disease. Beyond its application in fundamental research, NMR offers translational potential in clinical diagnostics and therapeutic monitoring.
By enabling the simultaneous and quantitative analysis of a broad range of metabolites in cell systems, tissues and biofluids, NMR-based metabolomics delivers a comprehensive and dynamic view of metabolism. 1D 1H NMR is typically used for fingerprinting, but in combination with 2D 1H-1H and 1H-13C NMR detailed information for the identification and quantification of metabolites becomes feasible. The emergence of public spectral libraries, signal processing algorithms, and innovative pulse sequences, allows for the deconvolution of a myriad of metabolites, directly from complex mixtures, such as biological samples. NMR spectroscopy can then be used in biomedical applications, identifying metabolic alterations associated with altered metabolic conditions (e.g metabolic syndrome, cancer, ageing, etc), or in pharmacological interventions. While NMR spectroscopy and MRI share a common physical foundation in nuclear magnetic resonance, their applications diverge significantly. Integrating NMR spectroscopy into clinical workflows offers a powerful, non-invasive avenue for metabolic assessment—bridging the gap between research laboratories and clinical practice, and paving the way for personalized medicine.
Sofia MOCO (Amsterdam, The Netherlands)
|
Espace Vieux-Port |
|
"Thursday 09 October"
Added to your list of favorites
Deleted from your list of favorites
C10
11:00 - 12:00
FT1-3 AUXILIARY HARDWARE
How to get the most out of your system ?
FT1: Cycle of Technology
11:00 - 12:00
Field Monitoring: Field cameras for higher performing systems.
Caroline LE STER (Keynote Speaker, Saclay, France)
11:00 - 12:00
Hardware for motion.
Adam VAN NIEKERK (Keynote Speaker, South Africa)
11:00 - 12:00
Hardware for physiology (incl. Pilot tones - promises and limitations).
Christopher ROY (Keynote Speaker, Lausanne, Switzerland)
|
Salle Major |
|
"Thursday 09 October"
Added to your list of favorites
Deleted from your list of favorites
D10
11:00 - 12:00
MRI TOGETHER
|
Salle 120 |
12:00 |
"Thursday 09 October"
Added to your list of favorites
Deleted from your list of favorites
A14
12:00 - 13:00
LUNCH SYMPOSIUM
|
Auditorium 900 |
|
LUNCH BREAK & LUNCH SYMPOSIUM
|
13:30 |
"Thursday 09 October"
Added to your list of favorites
Deleted from your list of favorites
A15
13:30 - 15:00
ET1-3 GRANT WRITING
ET1: Cycle of Research
13:30 - 15:00
How to win an ERC grant.
Andrada IANUS (Keynote Speaker, United Kingdom)
13:30 - 15:00
The 3 keys to writing a grant application.
David KARLIN
|
Auditorium 900 |
|
"Thursday 09 October"
Added to your list of favorites
Deleted from your list of favorites
B11
13:30 - 15:00
FT3-1 HARMONIZING RESEARCH
FT3: Cycle of Quality
13:30 - 15:00
Harmonization between scanners.
Qingping CHEN (Keynote Speaker, Germany)
13:30 - 15:00
Harmonization of AI-driven algorithms.
Bertrand THIRION (researcher) (Keynote Speaker, Saclay, France)
13:30 - 15:00
Harmonization of statistical-driven algorithms.
Shaun WARRINGTON (Keynote Speaker, Nottingham, United Kingdom)
|
Espace Vieux-Port |
|
"Thursday 09 October"
Added to your list of favorites
Deleted from your list of favorites
C11
13:30 - 15:00
FT1 LT - Smarter Sensing
Technological advances in MRI acquisition and contrast design
FT1: Cycle of Technology
13:30 - 13:32
#47700 - PG061 epgpy: an easy-to-use python library for MRI simulations.
PG061 epgpy: an easy-to-use python library for MRI simulations.
Whether it be for providing an insight into the logics of an MRI sequence, or for taking part into data fitting algorithms, numerical signal simulations play an important role in today’s research in the field of MRI. Typical cases include NMR signal fitting for parameter mapping, dictionary generation for MR fingerprinting and sequence optimization.
The extended phase-graph [1] (EPG) formalism provides an efficient alternative to classical isochromats sampling in Bloch equations simulations. Since its introduction, it was extended to model additional NMR properties (diffusion, magnetization transfer, 3D gradients).
Over the years, a number of simulation libraries have been developed, such as Jemris [2] and MR-zero [3], with different focuses (computational efficiency, sequence building) implementations (C++, python) and formalisms (Bloch, EPG).
Building on an original python implementation of the classical EPG operators, we implemented several EPG extensions and functionalities into epgpy, a simple to use and lightweight python library suitable for many use cases.
Epgpy is a pure python library, depending on the numerical library numpy and optionally on cupy for GPU calculations. Most functions and operators have unittests, and several examples reproducing published works are provided along with the source code.
[Basics] In epgpy, sequences are built as lists of operator objects. There are three basic classical EPG operators: E – the evolution operator, simulating the relaxation and precession of the magnetization – T – the transition operator, simulating instantaneous RF pulses – and S – the shift operator, for simulating phase states changes, such as induced by gradients. A code example for simulating a multi-spin echo sequence (MSE) is given in Figure 1, and the generated output in Figure 2. Note that the signals corresponding to each flip angle and T2 value are generated simultaneously and stored into separate entries of the simulation output.
[Extensions] Beyond the classical EPG algorithm, several extensions were implemented into epgpy: anisotropic diffusion [4] (operator D), exchange and magnetization transfer [5] (operator X), arbitrary 3D gradients [6] and B0/R2* time-dependent dephasing. To account for the latter phenomena, a quantize-and-merge algorithm based on Gao et al. [6] was used in combination with a 4-dimensional coordinates array comprised of the 3D wavenumbers and the time accumulation of the phase states, achieving a formalism similar to that of Endres et al. [7]. This constitutes an alternative to sampled isochromats for simulating the multiple spatial-temporal pathways of the magnetization. As an example, reproducing an experiment from Endres et al. [7], a 2D imaging sequence was simulated on a 64 x 64 brain phantom [8] defined by its proton-density, T1, T2 and T2* parameter maps. In Figure 3, several simulations of this sequence are shown, with either the EPG or the isochromats/Bloch approaches. The computing times for these simulations (laptop, Intel Core i7, 32 Go RAM) illustrate the superior efficiency and precision of the EPG approach compared to isochromats sampling.
[Differentiation] The signal's derivatives are needed in various situations, such as sequence optimization and confidence interval calculation. In epgpy, we implemented 1st and 2nd order explicit differentiation for the three classical EPG operators. As an example, reproducing an experiment from Lee et al. [9], an MRF sequence was simulated with epgpy with 400 TRs and flip angles and optimized with an objective based on the Cramer-Rao bound for the magnitude, T1 and T2. For this calculation, the 1st and cross derivatives of the signal with respect to T1, T2, and the 400 flip angles and TRs are needed. The results of this optimization are shown in Figure 4, closely matching those of the original publication. In developing epgpy, the focus was made on ease of use. This explains several design choices such as the use of python as unique language, the "pythonic", concise syntax, sequence definitions as simple lists of operators and reduced number of dependencies (essentially numpy). This results in a convenient framework for fast prototyping or educational purposes.
However, these choices also impact the computing efficiency. Thanks to numpy’s vectorization, simultaneous simulation of many signals is efficient, making epgpy suitable for dictionary generations. Further acceleration can be achieved on a GPU with the optional dependency cupy. On the other hand, non-vectorized calculations, due to sequences with many operators or when requiring differentiation, can be less efficient than with alternative libraries implemented in C++ or using automatic differentiation. With few dependencies, and a simple syntax for sequence definitions, epgpy can be used for fast prototyping. Moreover, by leveraging numpy’s vectorization and cupy’s GPU capabilities, epgpy can be efficiently used in certain optimization pipelines.
Pierre-Yves BAUDIN (Paris)
13:32 - 13:34
#47727 - PG062 MRIBuilder: sequence programming using non-linear optimisation.
PG062 MRIBuilder: sequence programming using non-linear optimisation.
Defining MRI sequences requires setting the values for the timings and other properties of all the RF pulses, gradients, and ADC events within the sequence. Most sequence programming frameworks (i.e., all major vendors as well as open-source solutions like pulseq[1] or pypulseq) rely on a procedural programming framework to set these parameters. In essence, it consists of a function, which takes as input target summary metrics from the user (e.g., echo time) and produces as output the exact timings and other parameters for all of the sequence components (left in Figure 1). This approach typically requires a large number of if-statements to resolve any edge cases.
As an alternative, we propose that sequence programming can also be thought of as a constrained optimisation problem. In such an approach, the timings and other parameters of the sequence components are considered free variables. The sequence properties, scanner hardware limits, SAR limitations, etc. can be modelled as constraints on these variables. We then use a constrained, non-linear optimiser to estimate the sequence component parameters (right in Figure 1).
We present MRIBuilder as a prototype for the proposed sequence programming framework. MRIBuilder is implemented in Julia[2]. It can produce outputs using the Pulseq file format[1], which allows the output sequences to be run on MRI scanners across multiple vendors.
Figure 2 illustrates how to define a new sequence in MRIBuilder. In addition to a name, the sequence is mainly defined by the order in which the sequence components will be played out. At this stage we do not set any of the component timings or other parameters. Instead, we define any number of constraints or summary metrics as equations.
This sequence definition is a hierarchical process. Each of the sequence components used in step 2 in Figure 2 is already defined at an earlier stage with its own constraints and summary measures. For example, the excitation pulse would have a constraint that the total integral of the slice-select gradient between the excitation pulse itself and the end of the block should be zero. These sequence components can be reused across many different sequences. The sequence defined in Figure 2 in turn, could be embedded within a larger block that repeats this sequence for different slices or gradient orientations.
When the user provides values for the summary metrics, these will be added as additional constraints. Optionally, the user can also provide a target for the optimiser (e.g., maximise the b-value). Otherwise, the default target of minimising the sequence duration is used.
This target function and all of the constraints are then passed on as analytic equations to the IPOPT library[3], which is a non-linear optimiser allowing for non-linear constraints. We find that IPOPT can produce optimal sequences even under a large number of constraints (as long as there is a valid solution). Ordinal or integer parameters (e.g., number of side lobes of sinc pulse) are supported using the Juniper library[4]. Both of these optimisers are accessed through the JuMP.jl framework. Figure 3 illustrates how the user would interact with the diffusion-weighted spin echo sequence defined in Figure 2. When only the b-value is set, the target of minimising the sequence duration will cause the RF pulse and readouts to become instantaneous (Figure 3A). However, adding a target voxel size and FOV ensures that more realistic slice-selective RF pulses and EPI readouts are used (Figure 3B).
Instead of setting the b-value, we can also set the echo time and maximise the b-value (Figure 3C). Figures 3D-F illustrate other constraints and targets that can be set by the user, which, respectively, minimise the diffusion time (Δ), fix the gradient duration (δ) or minimise the gradient slew rate. For these example sequences, the optimisation takes a few seconds each. Supporting the wide range of use cases illustrated in Figure 3 would require a huge set of if-statements in the traditional, procedural sequence programming framework to cover all edge cases. While not all of these examples are necessarily useful, they do illustrate the benefit of the proposed framework, where the job of defining all sequence component parameters is left to a constrained optimisation rather than the developer.
A potential limitation of using an optimiser for defining sequence parameters is that the numerical optimisation will not give exact results, but only within a predefined numerical precision. This might give issues with any MRI sequences that are very sensitive to tiny shifts in the sequence component timings or other properties. By defining an MRI sequence as a set of analytical equations, we can estimate the timings and properties of the MRI sequence components using a constrained optimisation.
Michiel COTTAAR (Oxford, United Kingdom), Zhiyu ZHENG, Benjamin C. TENDLER, Karla MILLER, Saad JBABDI
13:34 - 13:36
#47962 - PG063 Vendor-Neutral FOV Positioning for Non-Cartesian MRI in the Pulseq Framework.
PG063 Vendor-Neutral FOV Positioning for Non-Cartesian MRI in the Pulseq Framework.
MRI allows for flexible positioning of imaging volumes, which is essential for clinical use. Traditionally, off-center scanning requires embedding frequency and phase offsets into the pulse sequence, in a sequence-specific manner, which may be prone to errors. To address this, Magnald and Wehrli(1) proposed an automated Bloch equation-based method for computing phase offsets. Building on their work, M. Shafiekhani et al(2). integrated this into the open-source Pulseq(3) framework, enabling Cartesian FOV positioning across diverse MRI systems. However, many modern techniques prefer non-Cartesian k-space trajectories (e.g., radial, spiral), which complicate FOV shifts due to varying gradients. This work extends the previous Pulseq implementation to support FOV positioning in non-Cartesian sequences, maintaining vendor neutrality and sequence independence.
Theory
MRI sequences in Pulseq are typically designed at the isocenter as the reference for phase evolution. When the object is shifted, phase behavior at any point must remain consistent to avoid artifacts(4-6). This is achieved by applying calculated frequency and phase offsets to the transmitter and receiver throughout the sequence. Using a Bloch equation–based model(1), the required phase offset teta(t) at any time point t is: (Figure 1, EQ.1)
Here, G(t) is the gradient at time t, ???? is the gyromagnetic ratio, and miu(t) is the zeroth gradient moment. Applying this dynamic phase shift across the sequence allows an FOV shift by vector ∆r, preserving phase evolution without changing sequence structure or timing.
Metgods
To implement FOV positioning, we introduced a new function ‘TransformFOV’ within Pulseq that applies the necessary phase and frequency offsets to all receiver and transmitter events in the sequence, based on a user-defined FOV shift and rotation, generating the modified sequence.
This function performs several key steps for each block in the sequence. First, it extracts the start times of existing RF and ADC events, then computes the gradient moment vector (miuX, miuY, miuZ) at these time points. Using Equation (2) (Figure 1, EQ.2), the phase offsets are calculated.
For RF and ADC events with constant gradient amplitudes, the gradients (GX, GY, GZ) are extracted and used in Equation (3) (Figure 1, EQ.3) to determine the corresponding frequency offsets.
For RF and ADC events with time-varying gradients, the frequency offsets are calculated based on the amplitude of the gradient at the center of the RF and ADC events, using Equation (3). In addition, for gradients varying during separately for RF or ADC events a residual phase modulation vector is defined. For RF pulses this residual phase is immediately added to the RF pulse shape. For ADC events the Pulseq format has been extended to store this phase modulation vector, which is then applied to the measured data before reconstruction. This approach generates a modified sequence with updated phase and frequency offsets for both RF and ADC events for FOV shift of ∆r. Figure 2 and 3 show the modified sequence diagram for radial GRE and spiral sequences, respectively. Figure 4 presents images of the ACR phantom acquired using a radial GRE sequence (TR = 20 ms, TE = 8 ms, FOV = 256×256 mm², resolution = 0.8×0.8 mm², Nr = 256, slice thickness = 3 mm, flip angle = 10°) for different FOV positioning methods. Part A shows the reference image obtained by imaging the phantom at the isocenter and applying an image-domain shift of ∆r = [48, 0, 0] mm. This reference is compared with images acquired using the original sequence shifted via the Pulseq interpreter’s frequency-based method and the modified sequence using our phase-based offsets. Part B presents results for shifts of 8, 32, and 48 mm in both x and y directions. Differences relative to the reference image are also shown. The phase-based method produces more accurate results with fewer artifacts, particularly at larger offsets, demonstrating improved performance over the current frequency-based approach. Discussion and Conclusion
We extended the Pulseq framework to enable FOV positioning for non-Cartesian MRI using a phase-based algorithm. This method applies dynamic frequency and phase offsets based on Bloch equation modeling, independent of gradient shapes or scanner vendor. Tests with radial GRE show accurate object shifts compared to image-domain references, with better fidelity than the frequency-based method currently available in the Pulseq interpreter, especially at larger offsets.
Mojtaba SHAFIEKHANI (Freiburg im Breisgau, Germany), Patrick HUCKER, Maxim ZAITSEV
13:36 - 13:38
#47782 - PG064 Boosting contrast without ground-truth data: a self-supervised pulse-sequence optimization for contrast maximization.
PG064 Boosting contrast without ground-truth data: a self-supervised pulse-sequence optimization for contrast maximization.
Developing pulse sequences that generate high contrast between tissues is essential for Magnetic Resonance Imaging (MRI). Recently, frameworks such as MRzero enabled optimizing pulse sequence parameters to enhance tissue contrast [1, 2, 3]. However, they rely on supervised learning, i.e. on optimization with respect to pre-defined target images. This approach limits the potential for reaching new, unexplored tissue contrasts.
Here, we propose a novel self-supervised, reference-free pulse-sequence optimization method that does not involve any pre-defined target images. To this aim, we introduce a reference-free contrast-promoting loss function.
Consider a pulse sequence with parameter vector θ that provides an image x. We aim to optimize θ to achieve high contrast between tissues, without any pre-defined target contrast or precise knowledge of tissue locations in the image. For demonstration, we maximize the contrast between white matter (WM) and gray matter (GM) in a gradient echo (GRE) T1-weighted brain scan.
Reference-free loss function:
Let $m^{(k)}$ denote the tissue probability maps where k is the tissue type (WM or GM). We estimate the distribution of pixel intensities for each tissue via Kernel Density Estimation (KDE) [4], $$p(u; x, m^{(k)}) = (\|m^{(k)}\|_{1})^{-1} \sum_{(i,j)} m_{i,j}^{(k)} \kappa(u - |x_{i,j}|)$$, where κ is a Gaussian kernel. For tissues k and l, we propose to maximize contrast by computing the loss $$d_{m^{(k)}, m^{(l)}}(x) = D\left(p(\cdot; x, m^{(k)}), p(\cdot; x, m^{(l)})\right)$$, where D is the symmetric Kullback-Leibler divergence [5]. To avoid maximizing the contrast between any tissue and the image’s background, denoted by "bg", we add a term that measures the loss between the combined region of all tissues (k and l) and the background. Our reference-free contrast-promoting loss function is hence, $$L(x) = -(1-\lambda)d_{m^{(k)}, m^{(l)}}(x) - \lambda d_{m^{(k)} + m^{(l)},\ m^{(\mathrm{bg})}}(x)$$ where λ∈(0,1). Our loss is smooth on x, and assuming that θ↦x is a twice-differentiable model, we use gradient descent for the optimization.
Derivative-free optimization with our reference-free loss:
To further allow flexibility, we optimize the pulse sequence without reliance on a differentiable model, using derivative-free optimization [2]. During the optimization, updates of θ are deployed to an MRI system, MRI are conducted, and the loss gradients are computed empirically from the acquired data.
While derivative-free optimization was used in [2], in a supervised manner, here we apply self-supervised derivative-free optimization, using our reference-free loss. To accelerate the process, instead of on-the-fly scans, we sampled the parameteric landscape $\{(\theta, x)\}_{\theta \in \mathbb{R}^K}$ in advance, and performed the optimization offline. We drew random parameter vectors $\{\theta^{(i)}\}_{i=1,2,...}$, deployed each on the MRI system, and acquired images $\{x^{(i)}\}_{i=1,...}$. We used a random search [9] on the discrete search space: we initialize a point $\theta \in \{\theta^{(i)}\}_{i=1,...}$, then for each iteration, a candidate point $\theta_{c}$ is drawn near θ from a given probability distribution. If the image of $\theta_{c}$ has lower loss than that of θ, we update $\theta \leftarrow \theta_{c}$. Otherwise, no update is done. We demonstrate our self-supervised reference-free optimization method in 3 experiments, where we optimize the flip angle (FA), repetition time (TR), and echo time (TE) of a GRE sequence initialized with random parameters.
1. Numerical simulations: We simulated acquisitions of the BrainWeb digital phantoms [7] using MRzero [3] and Phase Distribution Graphs [6]. Our aim was to maximize WM-GM contrast. We applied our optimization process and back-propagated its gradient through the simulation, and then through the sequence using Pulseq-zero [8]. the initial parameters were FA of 5.9°, TR 19ms, and TE 13ms TE. The final values were FA of 7.1°, TR 54ms, and TE 48ms. The results show an increase in WM-GM contrast (Fig. 2).
2. Phantom experiment: We scanned a phantom consisting of vials with conditioner, cream, and yogurt using a 3T MRI system (Siemens, Prisma). Here we optimized the pulse sequence to maximize the contrast between two vials. The results show a notable contrast improvement (Fig. 3).
3. In-vivo experiment: We also demonstrate the method via in-vivo human brain scans, where we aimed to maximize the WM-GM contrast. The initial WM-GM contrast was very low, and the optimization process led to a much higher contrast (Fig. 4). Discussion and Conclusions:
We introduced a method for self-supervised reference-free pulse sequence optimization, which enables achieving high tissue contrast without any pre-defined target. It overcomes the limitation of supervised learning and can hence enable achieving novel contrasts. While demonstrated here for brain scans, our approach is very general and can be easily extended to other types of scans.
Alon GRANEK (Haifa, Israel), Efrat SHIMRON
13:38 - 13:40
#47961 - PG065 Simulation-Based Optimization of SPGR MRI Acquisition Protocols for Brain Contrast Similarity at 0.55T and 1.5T.
PG065 Simulation-Based Optimization of SPGR MRI Acquisition Protocols for Brain Contrast Similarity at 0.55T and 1.5T.
MRI is the gold standard for many clinical and diagnostic scenarios, but high associated costs limit accessibility [1]. MRI Research at low-field (LF) and ultra-low-field (ULF) is more attractive than ever [2], but they lack standardization in field strength [3],[4],[5]. This diversity complicates conventional interpretation and scanner operation as LF and ULF images are intrinsically different due to the variation in contrasts and the signal-to-noise ratio (SNR) [6]. While some sequences have been manually optimized or adapted from their HF counterparts [7] or even developed from scratch [8], this process can be long and expensive. In parallel, Deep Learning methods were proposed for image enhancement [9][10], but they require large datasets and can produce unreliable results, which is why a sequence-optimization approach is preferred. Thus, optimizing HF sequences for LF and ULF settings is necessary to accelerate the adoption of LF and ULF MRI. In this work, we study two algorithms to optimize 1.5 T sequence parameters for 0.55 T conditions, i.e., changed contrasts and less signal intensity, using simulations and a digital brain phantom.
Simulations were conducted using KomaMRI [11], which uses the Bloch equations. To reduce aliasing, we modified the conventional pipeline (Fig. 1) by identifying the unique spins of a digital phantom, creating unique-spins phantoms to make the simulator more responsive to spoiling effects and reduce the aliasing effect. The MR signals of the unique spins and their respective replicas are convolved with the phantom geometry's Fourier transform. The final image was obtained via the inverse Fourier transform.
The relaxation values of T1 and T2 at 0.55T were obtained from [12]. We used the brain phantom proposed in [14] with three tissues (white matter, gray matter, and CSF) and adjusted the proton density of low-field spins by 0.55/1.5. Noise was added in k-space data using a normal distribution. We assume that the noise is the same for HF and LF data.
We used a conventional RF-spoiled Gradient Echo (FOV = 0.35 m x 0.35 m, Nx = Ny = 128, NSA = 2, ADC sampling time = 0.512 ms, RF envelope = sinc, quadratic phases for RF-spoiling). The parameters to be optimized were the flip angle, TE, TR, BW, Ny, and NSA. The optimization problem minimized the MSE between a scaled LF image and the HF image, subject to timing constraints and coherence between conditions (Fig. 2).
We used the Frank-Wolfe (FW) and Nelder-Mead (NM) algorithms to optimize the parameters. It should be noted that the FW algorithm allows the optimization to stay in the parameters' manifold and introduce constraints, but the NM algorithm relies on simplex-like evaluations without gradients. Nevertheless, due to their numerical nature, the algorithms can still produce or attempt to evaluate invalid parameters; iterations were stopped when this happened. If no invalid parameters were provided, the number of iterations was limited to 30. Three T1w and three T2w sequences (see Fig. 3 and Fig. 4) were optimized using FW and NM algorithms. We noted that joint optimization of all the parameters did not converge. Thus, we optimized in two steps: first, flip angle, TE, TR, and BW; then, TR, Ny, and NSA. Results show that TR, Ny, and NSA may not require any optimization since MSE drastically decreases to its minimum with the first step. All the best parameters are found with FW. NM reduces the MSE, but not that effectively. It should be noted that the simulation approach allowed us to acquire an image from a digital phantom with 1.2 million spins and three tissues in less than a second. The results of the optimization are positive, having the fundamental parameters (flip angle, TE, TR, BW) changed as expected. Interestingly, it is possible to note that FW results are better than NM. This may be due to the geometrical information the gradient captures when optimizing. Moreover, FW significantly reduces the BW, which is coherent with increasing the SNR, however, this may produce exposure to undesired signals. Our approach assumes that the noise is the same in both HF and LF settings, however, this is limiting since it is well known that at ULF and LF, the predominant noise source could be the body. Also, by design, the simulations assume the same hardware constraints for HF and LF. While this is not entirely realistic, previous work such as [13] managed to use high-performance hardware. Besides, recent advances in MRI hardware could provide enhanced features to LF and ULF scanners. The optimization of acquisition parameters has been a topic of interest since the beginning of the MRI field. In this work, we demonstrate the feasibility of using numerical optimizers for the task in simulation settings. While our results are realistic, they still need to be validated in real MR scanners. These results can be considered the starting point for the experiential adaptation or optimization of sequences.
Guillermo SAHONERO ALVAREZ (Santiago de Chile, Chile), Ronal CORONADO, Carlos CASTILLO-PASSI, Marcelo ANDIA, Pablo IRARRAZAVAL
13:40 - 13:42
#47840 - PG066 End-to-End Optimization of Variable Flip Angle Schemes for Enhanced Apparent Resolution in 7T 3D Fast Spin Echo MRI.
PG066 End-to-End Optimization of Variable Flip Angle Schemes for Enhanced Apparent Resolution in 7T 3D Fast Spin Echo MRI.
Fast-spin echo (FSE) sequences are the backbone of time-efficient clinical MRI. To counteract the T2 decay during signal acquisition, variable flip angle (VFA) schemes [1,2,3] are typically employed which have an impact on the image quality and contrast and the specific absorption rate (SAR). At clinical ultra-high field (7T), however, dedicated approaches are necessary to manage the enhanced T2 decay and SAR. To this end, an alternative framework for VFA calculation [4] has been presented which is based on an end-to-end learning approach. The framework takes the k-space sampling into account and allows for flexible solutions regarding PSF, SNR, contrast and/or SAR. In this work, the framework is employed in three clinical sequences to showcase its potential to enhance the apparent resolution in different imaging scenarios.
Three test cases were considered with varying resolution- and imaging settings:
Test case 1: FLAIR with pTx [5] (resolution=0.45x0.45x0.45 mm³, interpolated to double resolution from the acquired data in each direction)
TR=8000ms, ESP=4.04ms, TEeff=300.0 ms, TI= 2250 ms, FOV=230 x 230 x 180 mm³, ETL=220, rel. SAR=6.0%, bandwidth=651 Hz/pixel, linear reordering scheme, acceleration via CAIPIRINHA sampling [6] (total factor 6, PE: 3, 3D: 2, shift: 1), Tacq=5.54 min.
Test case 2: FLAIR with pTx (resolution=0.7x0.7x0.7 mm³)
TR=8000ms, ESP=4.28ms, TEeff=304.0ms, TI= 2250ms, FOV=180 x 225 x 180 mm³, ETL=180, rel. SAR=7.4%, bandwidth=349Hz/pixel, linear reordering scheme, acceleration via CAIPIRINHA sampling (total factor 6, PE: 3, 3D: 2, shift: 1), Tacq=8.18 min.
Test case 3: T2 with pTx (resolution= 0.4 x 0.4 x 0.4 mm³)
TR=5000ms, ESP=6.01ms, TEeff=323.0ms, FOV=195 x 145 x 180 mm³, ETL=200, rel. SAR=5.1%, bandwidth=751Hz/pixel, linear reordering scheme, acceleration with compressed sensing sampling (total factor 6), Tacq=15.37 min.
Test cases 1 and 3 were optimized for solely PSF, while test case 2 was optimized for PSF- and SNR- trade off (see [4] for details). In vivo measurements were performed on healthy volunteers using a 7T whole-body MR system (Magnetom Terra.X, Siemens Healthineers, Erlangen, Germany) with a commercially available 8Tx/32Rx RF head coil (Nova Medical, Wilmington, MA). All MRI scans were under approval of the local ethics board, and were performed after written informed consent was obtained. For test cases 1 and 2, co-registration relative to a 3D MPRAGE dataset and bias field correction [7] were applied. All datasets underwent deep learning-based reconstruction [8]. To obtain the target signal for standard VFA calculation [2,3], WM was assigned T1=1500 ms and T2=50 ms. Fig. 1 shows the standard and optimized VFA schemes for all test cases. PSF-optimized VFAs (A, C) result in signal responses with strongly reduced PSF-sidelobes, while trade-off optimization (B) shows elevated signal responses at the echoes assigned to the k-space center. Fig.2 A depicts exemplary axial views of test case 1 with conventional reconstruction (CAIPIRINHA). The optimized VFAs result in improved delineation of WM/GM edges (green arrow) and enhanced visibility of smaller structures (orange arrow) when compared to a MPRAGE. Furthermore, bright spots which are typically located at the gyri edges and may be misinterpreted as “pseudo-lesions”, are strongly suppressed in the optimization when compared to the standard. Fig. 2 B depicts the deep-learning reconstruction, in which all features of the optimization are preserved.
Test case 2 with higher acquired resolution in comparison to test case 1 is shown in Fig. 3, where the improved visibility of smaller WM/GM structures results in better resolved structures in the putamen (green arrow).
Fig. 4 shows the high-resolution T2 images in test case 3 in coronal view. In this imaging scenario, the optimized images also benefit from the reduced PSF-sidelobes by revealing less apparent blurring in the WM/GM structures (e.g, green arrow), which is also seen in the hippocampal region. The visual and apparent resolution in 3D FSE sequences with very long echo trains often deviates from the nominal resolution due to PSF-broadening/contamination. The proposed framework mitigates this issue by explicitly optimizing for the PSF of multiple tissues (e.g., WM/GM in FLAIR and additionally CSF in T2-weighting). However, the PSF-optimization often results in decrease of baseline SNR (Fig. 2 A), which can be mitigated using advanced deep learning reconstructions which preserve the advantages of the optimization. Furthermore, the framework allows for a PSF-SNR trade-off optimization, which leads to dedicated and non-intuitive VFA schemes (Fig. 1 B) and enhances the baseline SNR. The employed end-to-end VFA optimization framework allows to optimize the apparent resolution for various clinical imaging scenarios and is compatible with advanced reconstruction and post-processing modules.
Peter DAWOOD (Gerbrunn, Germany), Martin BLAIMER, Angelika MENNECKE, Fraticelli LAURA, Weinmüller SIMON, Jonathan ENDRES, Peter M. JAKOB, Moritz ZAISS
13:42 - 13:44
#46723 - PG067 A protocol for free breathing 3D-CINE MRI with black blood contrast and fat suppression.
PG067 A protocol for free breathing 3D-CINE MRI with black blood contrast and fat suppression.
The gold standard for clinical assessment of ventricular volumes and myocardial masses for cardiac MRI is using 2D-CINE balanced SSFP (bSSFP) stacks, due to its high intrinsic blood pool to myocardium contrast and resulting accuracy (1,2). However, for many patients the multiple breath-holds needed for this assessment can be challenging and remains a limitation. Although free-breathing 3D-CINE MRI is available, at 3T it often suffers from inadequate blood-myocardium contrast (3,4)
Here we propose a novel approach for a free-breathing 3D-CINE acquisition and reconstruction with black-blood contrast and fat suppression (BB-CINE) performed at 3T without contrast administration. We hypothesize that blood-myocardium contrast is enhanced by our BB-CINE and visualization of cardiac structures is improved.
Our proposed approach is outlined in Fig. 1. The acquisition combines a free-running RF-spoiled gradient echo (GRE) sequence with an added preparation block consisting of blood and fat suppression at regular intervals (preparation duration = 24ms, total shot duration = 224ms). Fat suppression is achieved by a spectral presaturation with inversion recovery (SPIR) block (duration = 9ms), while blood suppression is achieved through an improved motion-sensitized-driven equilibrium (iMSDE) prepulse (velocity encoding = 30cm/s in FH, RL, and AP direction, duration 15ms)(5).
After each preparation block, GRE readouts begin at the central k-space point and then k-space is traversed in a pseudo-spiral manner on a Cartesian grid (6,7). The first three spirals can be seen in Fig. 1b.
Other sequence parameters include TR/TE=6.3ms/2.7ms and readouts per spiral arm=30. The size of the FOV is 268x420x100 mm3 (AP, LR, FH) with a reconstructed spatial resolution of 1x1x4mm3. The scan time is about 8 minutes.
Reconstruction utilized retrospectively sorting the data into a 5D matrix, including 15 cardiac bins based on the pulse oximeter signal and 4 respiratory bins based on k0 self-gating (8) or camera-based optical tracking. Respiratory motion is then corrected in image space through non-rigid registration of cardiac time-averaged images as previously reported (9). Image reconstruction is performed using the BART toolbox (10) using compressed sensing.
3 healthy volunteers (HV) underwent one scan session in which the described sequence was performed twice to assess the repeatability of measured volumes. Myocardium mass (including papillary muscles, obtained by multiplying the segmented volume by 1.05g/ml) and blood volume of the left ventricle were segmented using 3D Slicer (11) and values are reported for both scan and rescan as well as systolic and diastolic volumes. Cardiac frames for systole and diastole were chosen by visual assessment.
For comparison, for two HV additionally, a free-breathing 3D-CINE scan was performed based on a bSSFP sequence with bright blood contrast (BrB-CINE, imaging settings such as resolution and k-space sampling were tried to be kept as similar as possible to the BB-CINE scan). Reconstructed BB-CINE scans of two HVs are shown in Fig. 2. While for some cardiac bins blood suppression works well, for others it is not working perfectly; however, the blood signal predominantly is retained in the center of the heart, away from the edges and does not hinder contouring. Also, blurring of the myocardium can be seen in several bins.
While the here presented sequence allows for 3D segmentation of both myocardium and blood pool, segmentation was way more difficult in the acquired BrB-CINE scan due to the lower contrast between myocardium and blood pool as can be seen in Fig. 3.
The calculated systolic and diastolic blood pool volumes and myocardium mass for both scans for the three scanned HV can be seen in Fig. 4. Besides myocardium volumes for the third scanned volunteer segmented volumes are in good agreement for both scans. The proposed free-breathing BB-CINE technique enhances blood-myocardium contrast compared to BrB-CINE. While successful in segmenting cardiac volumes, some blurring and variability in blood suppression were noted. In conclusion, the BB-CINE technique offers a potential alternative to traditional cardiac MRI methods by enhancing blood-myocardium contrast without the need for breath-holding. While it shows potential for improved visualization and segmentation of cardiac structures, further optimization and validation are necessary to maximize its clinical utility.
Wilhelm STEHLING (Amsterdam, The Netherlands), Oliver GURNEY-CHAMPION, Kak Khee YEUNG, K.K., Aart NEDERVEEN, Pim VAN OOIJ, Eric SCHRAUBEN
13:44 - 13:46
#47213 - PG068 Pitfalls of using FLAIR for the imaging of the ventral dural lymphatic elements.
PG068 Pitfalls of using FLAIR for the imaging of the ventral dural lymphatic elements.
Impaired clearing of waste products from the brain via meningeal lymphatic vessels is a leading cause of many neurological disorders [1–3]. In humans and in marmoset monkeys, the meningeal lymphatics has been shown with contrast agent enhanced MRI [4]. Using MRI, efflux of a cerebrospinal fluid (CSF) tracer to the parasagittal dura was demonstrated in humans [5]. Imaging of the dural lymphatic system at high spatial resolution and without exogenous contrast agents would be highly relevant for diagnostic and clinical imaging of such structures. In a highly cited recent study, Albayram et al. describe hyperintense signal in the anterior cranial fossa on 3D T2-weighted fluid attenuated inversion recovery (FLAIR) scans without the administration of a contrast agent as “ventral dural lymphatic elements” [6]. The purpose of our study was to demonstrate that the bright signal observed on FLAIR is due to imperfect inversion as a result of strong field inhomogeneities in the anterior cranial fossa.
We replaced the conventional inversion pulse of FLAIR with an inversion pulse designed to be highly robust to inhomogeneities in the static magnetic field and the radiofrequency (RF) field. Robustness against field inhomogeneities was incorporated directly into the cost functional of an optimal control framework for pulse optimization [7], enforcing robustness to offsets in B0 for the range of ±2.4 ppm and RF robustness for a range of 80% to 115% of nominal B1 amplitude. Maximum available RF amplitude and phase variation were in compliance with scanner’s RF coil and specific absorption rate (SAR) limitations. The final optimized RF pulse had a duration of 2.01 ms. No other changes were made to the sequence. Data were acquired and compared for both the conventional FLAIR and the FLAIR with robust inversion in a phantom and in healthy volunteers. For the phantom experiments, a cylinder (diameter = 1.5 cm, length = 9.5 cm) filled with a 0.05 mmol/ml gadolinium solution was placed inside of a cylindrical phantom (diameter = 13 cm, length = 17 cm). The phantom was placed inside of the scanner with the Gadolinium-filled internal cylinder perpendicular to B0, in order for the paramagnetic solution to create field inhomogeneities around the cylinder. Scan parameters were: sagittal 3D acquisition with inversion time TI = 2400 ms in humans and 1650 ms in the phantom, repetition time TR = 8000 ms, voxel size acquired at 1.2 mm × 1.2 mm × 1.2 mm and reconstructed to 0.67 mm × 0.67 mm × 0.67 mm, field of view = 256 mm × 256 mm × 170 mm. The new inversion pulse exhibits nearly perfect inversion across the inhomogeneity phantom (Fig. B), whereas the conventional pulse results in bright artifactual signal in areas with strong off-resonance caused by the paramagnetic cylinder (Fig. A). In the human participant, FLAIR acquired with the conventional scan (Fig. C), as routinely used in clinical protocols, exhibits bright signal in the anterior cranial fossa. This hyperintensity is absent when using FLAIR with the highly robust inversion pulse (Fig. D). Notably, with the conventional inversion pulse, we obtain a result similar to that shown in the aforementioned paper by Albayram et al. [6]. Bright signal previously attributed to be lymphatic tissue is only visible with the conventional inversion pulse and likely artifactual in nature. It has been proposed that the brightness may be a flow artifact [8]. However, flow artifacts are usually absent from 3D FLAIR, where the inversion pulse inverts spins over a very large volume. Our results do not support the interpretation of hyperintense FLAIR signal in the anterior cranial fossa as ventral dural lymphatic elements. Rather, they provide evidence that the bright signal is due to incomplete inversion of CSF.
Christina GRAF (Vancouver, Canada), Alexander JAFFRAY, Armin RUND, Stefan STEINERBERGER, David LI, Alexander RAUSCHER
13:46 - 13:48
#47326 - PG069 Arbitrary center echoes in 3D RARE with offset concentric rings.
PG069 Arbitrary center echoes in 3D RARE with offset concentric rings.
The use of 3D RARE [1] sequences (Fast/Turbo spin echo) is ubiquitous in clinical MRI. With two phase encoding axes, multiple shots, and long echo trains, there are many view ordering possibilities.
Busse et al has described linear and centric view orderings for T2 and T1/PD weighted 3D RARE acquisitions, where the center echo implicitly follows from the prescribed k-space coverage and the echo train length (ETL) [2]. While vendors enable manual selection of effective TE, there is a lack of published information on how it's implemented. This highlights the need for view ordering techniques with arbitrary center echoes, especially with the emergence of research frameworks for pulse sequence programming like KS Foundation and Pulseq [3].
This abstract introduces CROC (Concentric Rings with Offset Center), a method for distributing echoes in 3D RARE imaging that enables an arbitrary center echo while smoothly spreading echo times across k-space. CROC is compared against circularly shifted linear view ordering which is currently used in Pulseq.
In CROC, echoes are assigned radially in the ky/kz plane, as illustrated in Figure 1A, but offset to achieve a later center echo. If the offset reaches the boundary of the prescribed k-space, the rings are stretched into ellipses to further increase the center echo.
Each echo segment (i.e. color in Figure 1A) is sorted azimuthally before shot assignment. This results in a radar-like sweep across k-space, as illustrated in Figure 1B. To avoid a singular point in which shots from the whole scan originate, the shots for the first echo are distributed linearly.
CROC was implemented in a 3D RARE sequence using the KS Foundation framework. The waveforms for a single shot is shown in Figure 2. Scans were performed on a 3T Signa Premier (GE Healthcare).
Phantom scans were acquired with CROC and circular shift with TR/TEeff = 2100/80 ms, ETL 96, matrix size 256×256×200, Isotropic voxels 0.8 mm, TRAPS flip angle modulation [4].
The in vivo scans were acquired with 2×2 2D CAIPIRINHA acceleration. TR/TEeff = 3000/93 ms, ETL 125, 384×384×150, Isotropic voxels 0.6 mm, TRAPS flip angle modulation. Figure 3 shows phantom and in-vivo images. Circular shift demonstrates significant ringing artifacts, whereas CROC effectively avoids these artifacts, resulting in a visibly sharper image. The offset is found with a binary search algorithm, making CROC agnostic to the acquired coordinates. This versatility allows CROC to be compatible with arbitrary sampling schemes, such as parallel imaging, 2D CAIPIRINHA [5], or Poisson disc sampling [6].
Ringing artifacts present with circular shift are removed with CROC, which is attributed to the avoidance of sharp transitions in the distribution of echoes. There is also a clear improvement in overall sharpness which we attribute to reduced T2 blurring. CROC effectively reduces ringing artifacts and improves image sharpness in 3D RARE imaging by providing a smooth distribution of echo times while also enabling arbitrary center echoes. This method offers versatility for various undersampling schemes and presents a significant improvement over circular shift view ordering.
Henric RYDÉN (Stockholm, Sweden), Ola NORBECK, Sophie SCHAUMAN, Mikael SKORPIL
13:48 - 13:50
#47947 - PG070 2D balanced steady state free precession deuterium metabolic imaging at 7T in vivo and on a head shaped phantom.
PG070 2D balanced steady state free precession deuterium metabolic imaging at 7T in vivo and on a head shaped phantom.
Deuterium metabolic imaging (DMI) is a powerful technique for tracking glucose metabolism and shows promise for studying tumor metabolism and assessing therapy by monitoring glucose, glutamine/glutamate, lactate, and fumarate [1-3]. The most standard DMI sequence follows a chemical shift imaging scheme [5,6]. Recently, a promising DMI sequence has been introduced based on a multi echo balanced steady state free precession (ME-bSSFP) pattern at high field (>7T) on small animals [5,6]. We demonstrate using a ME-bSSFP sequence at 7T, the capability to spectrally resolve metabolites, supporting DMI’s potential application in tumor progression treatment monitoring.
Hardware: All experiments were done on a Magnetom Terra 7T MRI (Siemens Healthcare, Erlangen, Germany) with a 15-element transverse electromagnetic mode (TEM) resonator for 2H transmit, a 16-element loop array for 2H receive, and a 4-element loop array for 1H transmit and receive (Virtumed LLC, Minneapolis, MN).
Materials: A custom head-shaped phantom was fabricated based on models from [7,8]. It was 3D printed using polylactic acid (PLA) at 80% infill density and coated with PlastiDip, sealed using OB1 adhesive for waterproofing [8] and filled with D2O at 0.1M and 0.1mM Dotarem to mimic physiological T₁. Sub compartments were filled with Sodium L-lactate (3,3,3-D₃), Fumaric acid (2,3-D₂) from CK Isotopes. NaCl was added to each compartment to achieve 1.0 S/m conductivity [9].
Sequence development: Sequence optimization simulations were performed in MATLAB (R2023b MathWorks) using equations from [5,10]. A 2D ME-bSSFP sequence was developed based on Siemens product Cine Vascular sequence framework with parameters matched to simulation results subject to SAR and timing limitations.
Phantom experiments: 2D ME-bSSFP was run with 23.36ms TR, 5 echoes starting from 4.01ms, 3.6 ms ΔTE, 7.3°flip angle, over a 64x64 matrix, 220 mm FOV, a 31.5mm slice thickness and 400 averages, resulting in a total acquisition time of 10 minutes and 13 seconds.
In vivo experiments: One healthy volunteer gave written consent and was recruited in accordance with ethical approval. To replicate in vitro experiments, D-Glucose (6,6-D₂) (CK Isotopes) dissolved in DI water was attached on the volunteer’s head. Volunteer was scanned immediately after drinking 15mL of D₂O. 2D ME-bSSFP was run with 8.5° flip angle, 16×16 matrix, 220 mm FOV, 31.5 mm slice thickness, and 600 averages, totaling 6 min 31 s acquisition time.
Data processing: Data was processed in MATLAB R2023b. WSVD coil combination was performed [12]. Magnitude and phase images were extracted and input into a modified Iterative decomposition of water and fat with echo asymmetry and least-squares estimation (IDEAL) fitting pipeline as described in Peters et al. [5, 11]. Metabolite maps for HDO, lactate, fumarate, and glucose were computed. 2D ME-bSSFP sequence was implemented on a 7T MRI scanner to enable spectral resolution of key deuterated metabolites. A realistic, anatomically shaped 3D head phantom was custom designed to fit the RF coil and mimic in vivo anatomy and metabolites (Figure 1). The ME-bSSFP sequence, employing five echoes was optimized for key metabolites (Figure 2). The 2D ME-bSSFP was implemented on the phantom and resolved signals from fumarate, lactate, and HDO (Figure 3). Preliminary in vivo studies in a healthy volunteer demonstrated the ability to resolve HDO and glucose (Figure 4). The successful resolution of fumarate, lactate, and HDO in the head-shaped phantom validates the ability of ME-bSSFP to separate relevant deuterated metabolites. Furthermore, the detection of HDO and glucose in vivo supports the feasibility of applying ME-bSSFP at 7T for metabolic imaging in human subjects. The metabolites resolved using this approach are highly relevant to tumor metabolism research, providing biomarkers for tumor progression and treatment efficacy through the assessment of metabolic activity and cell death [1,4]. Initial results are promising; however, further sequence optimization is necessary to accommodate scanner-specific technical constraints. The current SAR limitations restrict optimal flip angle from being achieved. Extending flip angle within SAR limits will boost bSSFP SNR further. Future work will focus on quantifying the SNR enhancement of ME-bSSFP relative to CSI and applying this technique in clinical studies with glioblastoma and Alzheimer's patient populations. ME-bSSFP is able to successfully resolve relevant metabolites making it a powerful tool in the future for DMI applications in the brain.
Acknowledgments: We thank Dr Frydman and Dr Peters for supplying the ME bSSFP simulation code. This research was supported by the NIHR Cambridge Biomedical Research Centre (NIHR203312) and the NIHR Applied Research Collaboration East of England. The views expressed are those of the author(s) and not necessarily those of the NIHR or the Department of Health and Social Care.
Sarah MILLER, James BROWNSEY, Masha NOVOSELOVA, Daniel ATKINSON, Jabrane KARKOURI (Cambridge, United Kingdom), Chris RODGERS
13:50 - 13:52
#46682 - PG071 3D Overhauser enhanced MRI of persistent nitroxide-labeled albumin contrast agents in living mice.
PG071 3D Overhauser enhanced MRI of persistent nitroxide-labeled albumin contrast agents in living mice.
The development of low-field MRI is an active area of research owing to cost efficiency and safety. Severe loss in sensitivity at low field can be alleviated by the use of dynamic nuclear polarization (DNP) through Overhauser effect in liquids [1] in the presence of free radicals. Thus molecular imaging purposes can be addressed with dedicated stable nitroxides, e.g. for proteolysis imaging [2]. In this context a new class of non-metal nitroxide-based macromolecular contrasts agent are being developed for conventional MRI [3]. Besides, pharmacokinetics of biodistribution of contrast agents is a relevant parameter to be assessed in vivo. Here it is proposed a remanent macromolecular system based upon a bovine serum albumin (BSA) grafted with a proxyl nitroxide spaced by a polyethylene glycol moiety in order to ensure nitroxide mobility. Overhauser-enhanced imaging at 0.19T in control mice in vivo and after a simple I.V. injection showed the persistence of the biocompatible contrast agent for hours in the blood system and several organs post intravenous injection. This study opens the way for biodistribution assessment of macromolecular probes in pathological situation like inflammation or cancer.
To keep the necessary mobility of the nitroxide, a bifunctional polyethylene glycol was synthetized from PEG and carboxy-PROXYL. The choice of a 5-carbon ring nitroxide ensures a slower rate of reduction in vivo. A succinymidyl amine-reactive group was added at the opposite end (Fig 1). This molecule was reacted with BSA in a 4/1 ratio. 200 µl of nitroxide-labeled BSA containing 40 mM PROXYL equivalent was injected into the tail vein of a mouse anesthetized with isofluorane (1.5-2% in air). MRI was performed at 0.194 T (MRI-Tech open-magnet) with gradients up to 20 mT/m (MRI-Tech, Canada Inc., Edmonton, Alberta). Electron spin saturation was achieved using a microwave cavity at 5443 MHz. The acquisition sequence was a fast spin echo (FSE) with the following parameters: repetition time (TR): 600 ms, echo time (TE): 9 ms, matrix: 64 x 32 x 32, field of view: 64 x 32 x 32 mm. To avoid heating, EPR saturation was only applied during acquisition of the central lines of the k-space. A control image (without EPR) was also acquired at each step. A phantom fiducial (3-carbamoyl proxyl tube of 1mM) was placed on the side of the mouse to serve as a reference for image signal, but also for spatial registration. Marked Overhauser effects were observed in large blood vessels and organs like gallbladder or spleen (Fig 2). Enhancements were in the range of -2 to -12 (i.e. 1200 % signal gain), taking into account the signal phase inversion pertaining to Overhauser effect in liquids though electron-proton dipolar coupling. Overhauser effect was observed for hours in vivo (Fig 3). Overhauser effect decreased in blood and spleen along time, which is in agreement with some bio-reduction and elimination processes. However, it increased in the gallbladder. The strong enhancement observed 6 hours post-injection indicated an active process of contrast agent elimination in the liver. Of note the overall remanence of the Overhauser effect indicated a significant resistance to bioreduction. This study demonstrates for the first time the occurrence of a long-lived Overhauser enhancement in vivo in the vascular system. This a first step towards the development of smart contrast agents a prolonged lifetime. Prolonged signal detection up to four hours post-injection underlines the robustness of the macromolecule-spacer-nitroxide system to biological degradation processes, which is particularly promising for the development of non-toxic probes for prolonged in vivo imaging. Beyond the enhanced permeability retention capacity of macromolecular systems for contrast generation, the upgrading of such systems with molecular targeting would constitute a considerable breakthrough for molecular imaging.
Work is in progress to increase the nitroxide concentration in order to visualize more regions of interest. Of course, tissue lesions might be attractive to investigate though this approach. It would also be interesting to target larger animals such as rats at ultra-low field (<10mT).
Joyce POKONG-TOUYAM (Bordeaux), Louis HOSPITAL, Eric THIAUDIÈRE, Philippe MELLET, Sylvain R.A. MARQUE, Gérard AUDRAN, Sophie THETIOT-LAURENT, Elodie PARZY
13:52 - 13:54
#47562 - PG072 Open-source diffusion-weighted bSSFP MRI pulse sequences to study the post-mortem brain using the GinkgoSequence development framework.
PG072 Open-source diffusion-weighted bSSFP MRI pulse sequences to study the post-mortem brain using the GinkgoSequence development framework.
Diffusion MRI (dMRI) allows us to investigate the brain's structural connectivity. Beyond the classical Pulsed Gradient Spin Echo[1] sequence that enables in-vivo studies with short acquisition time, Diffusion-Weighted Balanced Steady State Free Precession (DW-bSSFP)[2] sequences, although prone to motion artifact, show high SNR and strong diffusion sensitizations with unipolar diffusion weighting[3] and low flip angles, allowing for high-resolution dMRI data while keeping gradients from overheating. These sequences are particularly well suited in post-mortem studies at higher fields. However, most MRI manufacturers provide only a few dMRI pulse sequences, selected to cater to clinical applications. More advanced sequences can be shared within the community or adapted from proprietary source code, which can be challenging to obtain, modify, and lead to ownership conflicts.
Addressing these challenges, the open-source GinkgoSequence[4] framework makes state-of-the-art and custom pulse sequences programming accessible with a focus on dMRI applications: this work introduces a DW-bSSFP development suite in the GinkgoSequence framework.
Modular architecture: The GinkgoSequence framework has an object-oriented and modular architecture that limits code redundancy with a set of modules that can be picked and instantiated into a sequence (Fig.1). To ensure most sequences can be made, a complete panel of modules with mother classes that represent all the required stages coming into play in a sequence (preparation, excitation, encoding, refocusing, reading, and spoiling), and from which daughter classes are derived for specific needs, was implemented.
Specifically, DW-bSSFP sequences require unipolar diffusion weighting[3]. For that purpose, a new unipolar DW module was derived, as well as a new reading module to enforce the complementarity of gradients, required for balanced sequences. These modules were easily assembled (Fig.1) into 2D and 3D DW-bSSFP sequences, implementing options such as phase-cycling(PC)[5] to eliminate banding artefacts, and pauses between DW directions to enable system cooldown.
To assess their quality, these sequences were run on NeuroSpin’s 3T Prisma Siemens system with a post-mortem human brain hemisphere prepared by the iBrain Unit of Tours University, France. The acquisition protocol[6] included 30 DW directions with G=40mT/m and δ=13ms, 4 PC patterns applied on both DW and anatomical images, and 4 averages. The isotropic resolution was 1mm³ with TE/TR=20/40ms and a flip angle of 70°. The complete acquisition lasted 82 hours. Following our open-source approach, images were reconstructed using the Gadgetron[7] framework.
The analysis pipeline, performed using the Ginkgo toolbox[8], includes a reconstitution of the PC images, a computation of a precise mask of the brain, a Diffusion Tensor Imaging (DTI)[9] model enabling the mapping of fractional anisotropy and mean diffusivity quantitative maps and an analytical q-ball (QBI)[10] model to compute diffusion Orientation Distribution Function (ODF) maps, used by a streamline deterministic tractography[11] algorithm to obtain a whole-brain tractogram. Fibers were automatically labeled using the Ginkgo-Chauvel[12] and Ginkgo-Herlin atlases[13] for superficial and deep WM bundles. This pipeline ran on (1) PC and averaged DW data, (2) averaged but non-PC DW data, and (3) non-PC, non-averaged DW data for comparison. The acquisition and post-processing results can be found in Fig.2, displaying raw data and the main processing steps. ODF maps confirm the expected main orientations in each voxel, further supported by the tractogram and fiber labelling, giving the expected fiber bundles.
A comparison of results obtained for PC and non-PC DW can be seen in Fig.3, already displaying sufficient quality of averaged non-PC data. DW-bSSFP sequences, despite their remarkable sensitivity to the diffusion phenomenon, would not be well suited to investigate the brain microstructure in-vivo because of their long acquisition time and weakness to motion artefacts, restraining their use to post-mortem cases. The GinkgoSequence implementation is further limited by the framework: it is, for now, dedicated to Siemens MRI scanners and requires a research agreement with them to be compiled and used on machines. Implementing new modules to the GinkgoSequence framework allowed us to efficiently develop a customizable and open-source alternative to proprietary DW-bSSFP sequences, adding versatility to the variety of sequences already available within the framework. Their efficiency being shown by their success in detecting the state-of-the-art WM bundles, the sequences are envisioned to be used on post-mortem human brains with similar acquisition durations but higher spatial resolution due to the good quality of non-phase-cycled DW images, as well as in ultra-high field settings thanks to their unipolar diffusion mode and lower flip angles.
Eléa GRANIER (Gif-sur-Yvette), Anaïs ARTIGES, Christophe DESTRIEUX, Igor LIMA MALDONADO, Franck MAUCONDUIT, Cyril POUPON, Ivy USZYNSKI
13:54 - 13:56
#47676 - PG073 NMR relaxometry and transmembrane water exchange at low fields highlight the therapeutic potential of bumetanide drug in modulating glioblastoma invasion.
PG073 NMR relaxometry and transmembrane water exchange at low fields highlight the therapeutic potential of bumetanide drug in modulating glioblastoma invasion.
The 1H longitudinal relaxation rate (R1), measured at low fields by Fast Field Cycling NMR (FFC-NMR) technique, has been shown to distinguish between proliferation and invasion in glioblastoma (GBM) and has been correlated to transmembrane water exchange (t-Wex) mechanism [1-3]. Indeed, while water homeostasis is essential for all cells, cancer cells are particularly dependent on regulating this balance to support their rapid proliferation, invasion, and survival. Then, the extra/intracellular movement of water, driven by osmosis and metabolism, is contingent on pathological alterations in membrane transporter expression. The determination of the water intracellular residence time (τi) may be used as an early predictive biomarker of response to treatment. In this study, we investigate the bumetanide (BUM) as a repositioned drug affecting τi in a GBM mouse model. Originally used as a diuretic, BUM inhibits the NKCC1 transporter and has been linked to interactions with aquaporin channels AQP1 and AQP4, both of which are involved in GBM invasion [4-6].
In vitro measurements were performed on U87 and Glio6 GBM cell lines using Gd-based contrast agents (Gd-HPDO3A, 20 mm) with a 0.5 T benchtop NMR system (Stelar). Unlike Glio6 cells, which are characterised by an invasion/migration phenotype, U87 cells were treated with H₂O₂ (5 µM) to induce a similar invasion-like phenotype. Cells were incubated with BUM (1 µM) for approximately 17 hours before NMR experiments. In vivo measurements were performed on Glio6 mouse model obtained by injecting 4.10⁶ cells into the hind-limb muscle of immune-deficient nude mice (Athymic Nude-Foxn1nu, Envigo). BUM was administered intraperitoneally (5 mg/kg) every 12 hours during 14 days. NMRD profiles (R1 vs. magnetic field in the range of 0.12–23.5 mT) were acquired using a SpinMaster FFC2000 relaxometer (Stelar), following T2W 1H-MRI scans (Avance Neo AV300, Bruker) for tumour size assessment. The τi values were determined both in vitro and in vivo by modelling the bi-exponential magnetization recovery curve using the two-site exchange (2SX) model, as previously described [2]. In vitro, the BUM treatment caused an elongation of τi in both Glio6 and H2O2-stimulated U87 cell pellets (fig. 1). This was correlated to an increase of relaxation rate R1 measured at low field (0.01 MHz) as shown in [3]. In vivo, mice treated with BUM were divided into 2 groups based on tumour size: treated responders (R) and treated non-responders (nR). The effect of BUM on tumour size was most pronounced after six days of treatment and diminished thereafter (fig. 2A). Interestingly, in the R-treated group, we observed a BUM-induced effect on R1 values at low field (fig. 2B) and even more on R1tum (fig. 2C). R1tum was calculated as [R1-(1-X )R1h] /X with X, the tumour volume fraction derived from MRI and R1h, the R1 of the healthy leg. At 0.01 MHz, R1tum showed a significant difference between untreated and R-treated groups (p = 0.02). Figure 2D shows in vivo τi values measured on last day of treatment, when the tumor occupied at least 50% of the leg volume, corresponding to the threshold for acceptable NMR sensitivity. As expected, data reveal an increased τi in the R-treated group compared to the untreated group. In our previous work, we demonstrated the relevance of R1 measured at low field, linked to the t-Wex mechanism in cancer-related processes [1-3]. Specifically, we identified both R1 and τi as meaningful biomarkers of glioma cell invasion/migration. In the present study, we confirm these findings and demonstrate the sensitivity of these two biomarkers in detecting the therapeutic effects of drugs targeting water channels. Using BUM, our results highlight its potential to counteract GBM aggressiveness by impeding the invasion process. However, the therapeutic effect of BUM was not observed in all treated mice, likely due to variability in drug biodistribution. In future work, we plan to investigate the dose/response relationship of BUM and monitor its pharmacokinetics to correlate plasma drug levels with t-Wex modulation. This study confirms the value of FFC-NMR in detecting R1 at low fields and τi as in vivo biomarkers for monitoring tumour progression and treatment efficacy, Furthermore, it highlights BUM potential as a repositioned GBM therapy that may impede invasion by slowing the t-Wex mechanism. Furthermore, the importance of Field-Cycling Imaging (FCI, [7]) in visualising peritumour regions with infiltrating tumour cells (areas often undetectable with conventional imaging) is highlighted, due to its high sensitivity for monitoring response to t-Wex-targeted treatment.
Simona BARONI (Torino, Italy), Sahar RAKHSHAN, Michèle EL-ATIFI, Ayda ZARECHIAN SOUDANI, Lionel M BROCHE, François BERGER, Simonetta GENINATTI CRICH, Hana LAHRECH
13:56 - 13:58
#47908 - PG074 Exploring the Warburg effect in glioblastoma at 3 Tesla: A comparison of deuterium MRI with structural MRI and amide proton transfer-weighted imaging.
PG074 Exploring the Warburg effect in glioblastoma at 3 Tesla: A comparison of deuterium MRI with structural MRI and amide proton transfer-weighted imaging.
In glioblastoma, altered metabolism plays a crucial role in promoting upregulated cell proliferation [1]. Under normal conditions, cells predominantly metabolise glucose via oxidative phosphorylation. However, glioblastoma cells can favour glycolysis; this phenomenon, known as the Warburg effect, is hypothesised to facilitate uncontrolled cell proliferation [2,3]. Non-invasive assessment of tumour metabolism with imaging may offer valuable information on the metabolic phenotype [4], potentially enabling personalisation of therapeutic strategies and improved treatment-decision making. Deuterium metabolic imaging (DMI) is an MRI technique that enables in vivo visualisation of divergent metabolic pathways in the brain [5]. It can spatially quantify glycolysis by mapping levels of lactate (Lac), as well as oxidative metabolism through the combined signal of glutamate and glutamine (Glx). Decreased Glx or elevated Lac may indicate regions where the Warburg effect predominates. DMI shows promise for the field of neuro-oncology; however, the majority of in vivo research has been performed at experimental field strengths, and its validation at clinical field strengths is limited [6,7]. Here, we acquired DMI at 3 Tesla in patients with glioblastoma, and compared the metabolic pathways in different regions identified on structural MRI and amide proton transfer-weighted (APTw) imaging. The latter detects mobile proteins and peptides, reflecting regions with active cell proliferation [8].
Four patients with glioblastoma underwent DMI and APTw imaging alongside their clinical MRI-scan for radiotherapy (3T SIGNA Premier, GE Healthcare). Following a 6-hour fasting period, deuterated glucose was administered orally at a dose of 0.75 g/kg body weight (maximum dose 60g). A dual tuned birdcage head coil (Pulseteq, Cobham, Surrey, UK) was used to acquire a T1w reference image (0.5x0.5x1.0 mm3) and perform higher-order shimming to improve the magnetic field homogeneity. 60 minutes or more after glucose administration, 3D 2H-MR spectroscopic imaging (MRSI) was performed (FOV=30 cm; matrix=10x10x10; TR=250 ms; flip angle=60°; no. of excitations=1678; spectral points=700; bandwidth=5000 Hz; NEX=4; acquisition time=28:00 min), along with the acquisition of unlocalised MR spectra before and after MRSI. Metabolite maps were derived according to the methods of Khan et al. [9], included glucose (Gluc), Glx and Lac, and were upscaled to 20x20 via zero-filling. Nearest-neighbour interpolation of the normalised Glx/Gluc, Lac/Gluc and Lac/Glx maps was performed to match the matrix size to that of the T1w reference image. After DMI acquisition, we swapped the DMI birdcage coil with a 48-channel head coil to acquire the clinical MRI-scan and a 3D snapshot CEST-sequence (matrix=128x128, voxel size=1.7x1.7x3 mm3; frequency offsets: ±100, ±50, ±10, ±8, ±6, ±5, ±4, ±3.5, ±3, ±2.5, ±2, ±1.5, ±1.2, ±1, ±0.8, ±0.5, ±0.25, 0 ppm) to generate an APTw image as described by Wu et al [10,11]. Segmentations of the whole tumour, contrast-enhancing (CE) tumour, non-enhancing (NE) tumour and contralateral normal-appearing white matter (cNAWM) were created using the structural MRI-scans and registered to the T1w reference image of the DMI-scan. In addition, a biological tumour volume (BTV_APT) was segmented based on elevated APT signal [12]. Thereafter, we compared the Glx/Gluc, Lac/Gluc and Lac/Glx across segmented regions. The median Lac/Glx in the whole tumour, CE tumour, NE tumour and BTV_APT were 0.83 (IQR: 0.55-1.11), 0.79 (IQR 0.55-1.02), 0.84 (IQR 0.55-1.15) and 0.79 (IQR 0.55-1.04), respectively. In the cNAWM, the median Lac/Glx was 0.37 (IQR: 0.19-0.66). Fig. 1 shows a box plot of the Lac/Glx across different regions. In the tumour regions, the median Glx/Gluc ranged from 0.39 – 0.42 and the median Lac/Gluc ranged from 0.32 – 0.39. The median Glx/Gluc and Lac/Gluc in the cNAWM were 0.42 (IQR: 0.33-0.57) and 0.17 (IQR 0.08-0.31), respectively. Fig. 2 shows a box plot of the Glx/Gluc and Lac/Gluc. The Dice similarity coefficients between the whole tumour (based on structural MRI) and BTV_APT ranged from 0.47-0.51. In Fig. 3, the normalised metabolic maps and segmentations of a patient are shown as an example. Table 1 presents the median metabolic ratios per patient. We observed elevated Lac/Glx in tumour defined on structural MRI and APTw imaging, implying the presence of the Warburg effect. The higher median Lac/Glx in the tumour primarily arose from elevated Lac/Gluc as the median Glx/Gluc signal was similar across regions. The limited spatial resolution of DMI, and small sample size, warrant further investigation with a larger cohort. However, we observed notably higher Lac/Glx in the tumour region, indicating sufficient signal-to-noise at clinical field strength. Our work highlights the potential of DMI at 3 Tesla for non-invasive visualisation of the Warburg effect in human glioblastoma, further supporting its feasibility at clinical field strength.
Patrick L.y. TANG (Rotterdam, The Netherlands), Sebastian BAUER, Alixander S. KHAN, Esther A.h. WARNERT, Alejandra MÉNDEZ ROMERO, Marion SMITS, Edward J. PEAKE, Tomasz MATYS, Mary A. MCLEAN, Ferdia A. GALLAGHER
13:58 - 14:00
#46446 - PG075 Interleaved TMS-fMRI using BOLD and diffusion functional contrasts to investigate inhibitory neural activity.
PG075 Interleaved TMS-fMRI using BOLD and diffusion functional contrasts to investigate inhibitory neural activity.
Brain activity has been extensively studied using blood oxygen level-dependent (BOLD) contrast, which captures both positive (excitatory) and negative (potentially inhibitory[1]) signals. More recently, the apparent diffusion coefficient (ADC)-fMRI (neuromorphological coupling) has shown promise for improved spatial and temporal specificity compared to BOLD[2-4]. While excitatory activity has been reported as negative ADC response, the ADC counterpart to negative BOLD remains to be investigated. Transcranial magnetic stimulation (TMS)-fMRI can be used to modulate brain activity during fMRI acquisition. Previous studies have shown that subthreshold TMS (unilateral, 5 Hz) to primary motor cortex (M1) can induce contralateral M1 negative BOLD responses[5,6]. However, this approach remains highly experimental, with few prior studies[7]. Our study aims to replicate these BOLD findings and explore the corresponding diffusion fMRI (dfMRI) responses.
Data from four subjects were collected on a 3T MRI system with two 7-channel MRI receiver coils (7ch) and a TMS figure-eight coil (Fig. 1A). The protocol included five scans: anatomical bSSFP (one with the body coil –BC-, one with the 7ch coils), isotropic dw-SE-EPI (spherical b-tensor encoding[8]), multi-echo GE-EPI (T2*-BOLD), and T1 MPRAGE (64-channel coil). During dfMRI scans, alternating volumes at b=200 and 1000 s mm-2 were acquired. Acquisition parameters are shown in Fig. 1C. Pre-processing included: magnitude image denoising, Gibbs unringing, despiking, TOPUP and motion correction. BOLD echos were combined using Tedana. dfMRI provided b200, b1000 and ADC timecourses (calculated from pairs of b=200/1000 images). b200 and b1000 timecourses combine BOLD (via T2 weighting) and diffusion contrasts, and can be expected to be comparable to BOLD (minus direct blood contributions). We used 5 Hz TMS stimulation at 90% rest motor threshold (RMT, determined using electromyography) on the right M1, using MagVenture Magpro XP. The task consisted of six repetitions of stimulation blocks, where TMS pulses were interleaved with the MRI acquisition to avoid image artifacts (Fig. 1B). GLM analyses (FSL FEAT) were performed with temporal filtering (>0.01 Hz), nuisance regression (motion parameters and global signal), and cluster correction (z≥3.1, p<0.05), to detect positive or negative association with the task. The task was modelled as a boxcar function (dfMRI[2,3,9]) or convolved with the haemodynamic response function (BOLD). The z-score maps were registered to MNI space (functional→TRUFI BC→TRUFI 7ch →T1→MNI) for group analysis. Fig. 2 shows group-average z-score maps and timeseries averaged across significant voxels. In BOLD, positive z-scores are observed in the bilateral supplementary motor area (SMA) and inferior parietal lobule (IPL), and ipsilateral (relative to TMS stimulation) premotor cortex (PM). Negative z-scores appear in the contralateral M1, including its midline region. Both b1000 and b200 show positive z-scores in ipsilateral primary sensory cortex (S1). While b1000 shows positive z-scores in ipsilateral PM and negative in contralateral M1, b200 shows negative z-scores in contralateral S1 and superior parietal lobule (SPL). No ADC association with the TMS blocks is found in these regions. Fig. 3 highlights the between-subject variability in task-related activation. For instance, positive (sub-01 and 03) and negative (sub-02) z-scores are seen in SMA in BOLD. We replicated known BOLD patterns, such as negative BOLD in contralateral M1[5,6,10], with similar change in relative signal as in [10]. We found positive z-scores in SMA[5,10], with similar change in relative signal as in [10] at 110% RMT, while the opposite polarity was seen in one subject, as reported in [6]. Bilateral positive clusters in the lateral PM were also found in [5]. Overall, this suggests the experimental setup is reliable. Unlike [5,6,10], we observed ipsilateral M1/S1 activation, which may reflect an overestimation of the RMT and suprathreshold stimulation. We also reported activity in the IPL and SPL, known for sensory processing and sensorimotor integration[11,12]. Shared patterns between BOLD, b1000 and b200 maps demonstrate the setup applicability beyond BOLD contrast. The partial positive BOLD association in contralateral M1 or SMA is absent in b200 and b1000 and may reflect a direct blood contribution. Lower z-scores in ADC maps likely stem from lower contrast-to-noise ratio (CNR). Key challenges include minimising task-related motion, maintaining good SNR despite coil limitations, and ensuring precise stimulation, both in terms of location and intensity. The observed BOLD responses were broadly consistent with prior findings in contralateral M1 and bilateral SMA. For the first time, we report dfMRI responses to TMS, that partially overlap with BOLD patterns. However, due to the lower CNR of ADC-fMRI, a larger sample size is needed to robustly assess its sensitivity to neuromodulation.
Inès DE RIEDMATTEN (Lausanne, Switzerland), Arthur SPENCER, Roberto MARTUZZI, Vincent ROCHAS, Filip SZCZEPANKIEWICZ, Ileana JELESCU
14:00 - 14:02
#47692 - PG076 Highly accelerated snapshot readout for high-resolution CEST imaging.
PG076 Highly accelerated snapshot readout for high-resolution CEST imaging.
Many relevant structures for diagnosis in human brain are very small and can be distributed over the whole brain [1]. The detection of small metastases or epilieptic foli with the help of GluCEST [2] requires high-resolution CEST images. The cCEST approach [3] for creating different contrasts, however, requires a large number of images with identical readouts. To reduce the measurement time to a minimum and still obtain reliable data, the snapshot CEST method [4] allows only a very limited acquisition time. To cover the whole brain with a snapshot, we must encode the whole brain k-space with the same number of k-space lines, to avoid further signal decay. Recent developments in parallel imaging and deep learning-based denoising now make it possible to improve both coverage and the resolution in single snapshot readouts.
A newly developed snapshot gradient echo (GRE) CEST sequence allows the use of accelerating methods GRAPPA [5] and CAIPIRINIA [6]. It is also possible to further improve and accelerate these methods with deep learning-based techniques. A deep learning reconstruction similar to [7] was chosen that uses data consistency and denoising within each iteration step and is based upon the variational network approach [8]. This makes it possible to increase the acceleration in Phase Encoding direction (PE) to 4 and in Slice Encoding direction (SE) to 3, resulting in a total acceleration of 12 from GRAPPA 2 or 4 or CAIPIRINIA 6. Figure 1 lists the corresponding sequence parameters. The required reference scan is acquired once at the beginning of the measurement, which slightly increases the total measurement time but reduces the number of lines to be acquired after one CEST saturation. Moreover, Sinc interpolation was employed to double the resolution. This leads to a final resolution of 0.5x0.5x2 mm. The generated CEST contrasts were created and evaluated using the comprehensive CEST (cCEST) approach [3,9]. The B1 correction was performed using a DREAM sequence [10].
All experiments were performed on a 7T Terra.X VA60 (Siemens Healthineers) scanner and an 8Tx/32Rx head coil (Nova Medical) and under approval of a local ethics committee. Figure 2 shows a selection of cCEST contrasts in a sagittal slice, Figure 3 shows the corresponding contrasts in a transversal slice. The measurements were taken in different subjects. The whole brain sequence with a measured resolution of 2x2x4mm is already comparable to the existing cCEST snapshot measurement in terms of resolution quality. However, the coverage in the z-direction was almost tripled. The contrasts shown here 1µT Amides,1µT ssMT, 1µT NOE and 4µT Hydroxy show equivalent results for cCEST and whole brain. The high-resolution images show significantly improved resolution in Amides, ssMT and NOE. The contrast in the hydroxy is also significantly improved, but a clear increase in noise can be recognized here. The demand for high resolution whole brain coverage created the need for an improved CEST sequence. Since CEST imaging already requires a large amount of time due to the high number of offset images acquired, the acquisition had to be within the time window of a single snapshot with below 1000 k-space lines per shot [4]. This is possible thanks to the higher SNR in 7T UHF imaging, improved acceleration methods and deep learning-based denoising. CAIPIRINA 6 accelerated whole brain coverage provides stable results over the entire brain and has already become the internal standard. Furthermore, the highly accelerated high-resolution sequence with CAIPIRINIA 12 delivers astonishingly high-resolution images with only a marginal increase in noise. It must also be considered that the signal in the selective slice is measured in a slice only 2 mm thick. Whether this image quality remains consistent in different groups of people and patients still needs to be investigated further. The proposed gradient echo CEST sequence enables the integration of novel acceleration methods and deep learning based post-processing steps. Whole brain coverage can thus establish itself as the new standard in CEST imaging and it is even possible to increase in-plane resolution despite a significantly increased number of slices.
Acknowledgements:
Thanks to Moritz S. Fabian for providing the evaluation pipeline.
Funded by IDL@7T, BMBF.
Martin FREUDENSPRUNG (Erlangen, Germany), Patrick LIEBIG, Simon WEINMÜLLER, Magda DURATE, Moritz ZAISS
14:02 - 14:04
#47706 - PG077 Towards selective mr imaging of deuterium labelled fatty acids.
PG077 Towards selective mr imaging of deuterium labelled fatty acids.
The exchange of fatty acids between organs plays a crucial role in metabolic processes related to tumor energy metabolism [1] and BAT activation [2]. Deuterium metabolic imaging (DMI) of labeled fatty acids is a promising technique to investigate inter-organ energy transfer. We characterized the NMR properties of 31D-palmitic acid and developed a fatty acid-specific DMI strategy. Our findings show a super-fast T1 recovery and rapid T2 decay of 31D-palmitic acid, enabling selective DMI of fatty acids using ultra-short time to echo (UTE) imaging and short TR, as demonstrated in in-vivo imaging.
All measurements were carried out on a Bruker BioSpec 70/20 system and a 17 mm custom-build 2H T/R surface coil in combination with a 40 mm 1H T/R bodycoil. Animal experiments were carried out according to local animal welfare guidelines and were approved by local authorities. Relaxometry: 31D-palmitic acid was complexed to 10% bovine serum albumin (BSA) in 0,45% NaCl. For 2H NMR spectroscopy 1mM 31D-palmitic acid was prepared in a 5 mm NMR sample tube and analyzed for 2H- T1 relaxation rates with an inversion recovery experiment (IR = [2.7, 3.5, 6, 12, 20, 40, 60, 120, 200] ms, TR = 1000 ms, NEX = 1000, bw = 2 kHz, 256 points) and for T2 measurement a non-selective pulse acquire protocol (TR = 400 ms, NEX = 1000, bw = 2 kHz, 256 points) was used. Fitting was done with jMRUI package AMARES. To quantify spin-saturation by TR, again the 1mM 31D-palmitic acid sample was used. A non-selective pulse acquire protocol with gradient and RF spoiling, varying points based on available TR time, and a bandwidth of 3.2 kHz was used with TR = [3.7, 6, 11, 20, 30] ms.The number of averages were adopted to fill 4 min total scan time. Flip angle was kept at 90° at all experiments. All spectra were zero-padded to reach a final resolution of 128 points. Peak heights of HDO and CD2 were analyzed. In addition, deuterium imaging experiments at two different TR (= 3.7, 30 ms) were acquired from water, 1mM 31D-palmitic acid and sunflower oil filled into 5 mm NMR tubes. DMI pulse sequence: hard-pulse excited 2D-UTE TR = [3.7, 30] ms, TE = 1 ms, NEX = 650, bw = 50 kHz, alpha = 90°, matrix = 64 x 64, resolution = 2.5 mm x 2.5 mm, Taq = 4 min.
In-vivo DMI: 6-month-old C57Bl6/J mice were scanned in prone position. A 1-ml syringe was filled with 100 µl of BSA-complexed 31D-palmitic acid (1mM) and placed on top of the 2H coil and the coil was placed on top of interscapular brown adipose tissue. The DMI protocol was the same as for the imaging phantom study. Figure 1 (a, b) displays the data and fitted curves for T1 and T2 relaxation time measurements. The T1 of HDO in 1 mM 31D-palmitic acid (10% BSA) was 41.4 ms, and the T1 of CD2 was 10.5 ms (R² = 0.9614 and 0.9829). T2 fitting gave 26.9 ms for HDO and 2.46 ms for CD2 (R² = 0.9032). These results motivated further investigation into steady-state saturation of HDO to enhance the specificity of DMI for fatty acids. Spectra acquired under steady-state conditions are shown in Figure 2a. Additionally, the short T2 of the fatty acid signal necessitated the use of UTE readouts for DMI. Initial phantom images acquired at different TR values are presented in Figure 2b. Signal differences among water HDO, 1 mM 31D-palmitic acid (HDO + CD2), and sunflower oil (CD2) are summarized in Figure 2d, while the theoretical mM concentrations of deuterium signal-producing compounds are listed in Figure 2c. The clear separation between fatty acid samples and HDO prompted our first in-vivo measurements, shown in Figure 3. Remarkably, a strong contrast between pure HDO-containing tissues (e.g., muscle) and adipose tissues of various types was achieved by reducing the TR from 30 ms to 3.7 ms, the shortest TR achievable under the current readout conditions. Due to the inherently short T1 and the minimal chemical shift difference of approximately 200 Hz between HDO and CD2 at 7T, achieving spectrally selective saturation of HDO is challenging. Therefore, alternative methods like steady-state saturation were explored. As shown in the figure inset (Figure 1a), reducing the TR results in markedly greater suppression of HDO compared to the fatty acid signal. Interestingly, at TR = 3.7 ms, sunflower oil exhibited an even greater signal enhancement relative to HDO, with a ratio surpassing 1:3 (as opposed to the expected 1:1 spin-density), indicating an even shorter T1 time compared to 31D-palmitic acid. In-vivo measurements confirmed the fatty acid selective feature of the acquisition with HDO suppression sufficiently to discriminate between adipose tissue and muscle. This study demonstrates the short T1 of deuterium in fatty acids, facilitating selective imaging of fatty acids over the naturally abundant water signal. The integration of short TR with UTE sampling, yielding an acquisition time of 4 minutes, enables dynamic uptake studies of deuterium-labeled fatty acids, thereby advancing the application of DMI in lipid tracing.
Clemens DIWOKY (Graz, Austria), Martina SCHWEIGER
14:04 - 14:06
#45948 - PG078 Absolute quantification of muscle phosphorus metabolites using an external phantom.
PG078 Absolute quantification of muscle phosphorus metabolites using an external phantom.
Phosphorus MR spectroscopy (31P MRS) is very useful method for evaluation of muscle energetic metabolism. Besides metabolites observed in 31P MRS at rest, dynamic 31P MRS provides an estimate of mitochondrial capacity (Qmax) that is, however, also partly dependent on pseudo-normalization of individual signals relative to an assumed stable intracellular adenosine triphosphate (ATP) concentration of 8.2 mmol/l in skeletal muscle (1).
The aim of our study was to improve the diagnostic accuracy of 31P MRS by introducing absolute quantification using an external phantom and a volume coil.
Detection profile of our volume 1H/31P coil (Stark, Germany) was characterized using a large phantom with a defined phosphocreatine (PCr) concentration and a small phantom containing phosphonate (resonates on ~ 23 ppm) (2). The small phantom (designed to fit within voxel of CSI matrix, which also covers the entire calf muscle) was positioned inside the dual 1H/31P volume coil, using a custom 3D-printed holder (see Fig. 1).
The calculation of individual metabolite concentrations was done in two steps:
1) Signal intensities in the CSI voxels of the gastrocnemius (GM) and soleus (SM) muscles (see Fig.2) were calculated and compared with the phosphorus signal from the phantom to determine the concentration of PCr. We evaluated only PCr signal due to very good SNR and the reliability of its evaluation. Coil’s detection profile was also considered and included in the calculation.
2) Subsequently, using the individual ratios of signal intensities obtained from the 1H/31P surface coil (capturing primarily the signal from GM and SM), we calculated the concentrations of other 31P metabolites — Pi, ATP, and PDE — in GM and SM.
We conducted examinations on three groups of subjects: healthy volunteers (n=13, age=52.2±13.0 years), patients before and after liver transplantation (n=11, age=62.5±7.3 years) and patients with a history of diabetic foot syndrome or ongoing diabetic foot syndrome (n=9, age=60.4±9.1 years) at 3T VIDA scanner (Siemens, Germany).
Study was approved by local the Ethics Committee and informed consent was obtained from all subjects before entering the study.
Subjects underwent 31P MRS using surface 1H/31P coil both at rest (FID sequence with repetition time (TR)=15 s, bandwidth 3000 Hz, 16 acquisitions) and with exercise (FID sequences with TR=2 s, bandwidth 2000 Hz, 420 individual spectra each with 1 acquisition - 1 minute rest, 4 min exercise and 9 min. recovery (3)). In addition, 2D CSI (matrix 12 x 12, FOV 200 x 200 x 60 mm, TR/TE= 2000/2.3 ms, 10 averages, vector size 2048, bandwidth 3000 Hz) was measured with a volume 1H/31P coil for absolute metabolite quantification.
The data were evaluated using both the conventional approach (signal ratios relative to the sum of all signals, PCr/Pi ratio and Qmax based on pseudo-normalization) and absolute quantification (concentrations of PCr, Pi, ATP, and PDE in mmol/l, and Qmax recalculated). We compared both sets of results in terms of their ability to detect significant differences — considering the severity of the diagnoses, differences between the groups were expected to be significant. Statistical analysis was performed using ANOVA followed by Tukey’s post hoc test. A standard alpha level of 5% was used to assess statistical sign. The results are summarized in Table 1. Based on absolute quantification, ANOVA demonstrated significant differences between the groups of subjects in the concentrations of PCr, ATP, as well as in the Qmax value, significant difference in relative ratio was only found in ATP/Ptotal ratio.
Post hoc analysis revealed that group with diabetic foot syndrome exhibited significantly different values in these parameters compared to the other groups. It is evident that absolute quantification enabled the detection of significant changes in relatively small groups of subjects for certain metabolites at rest, as well as in the resulting Qmax value. In contrast, as relative ratios, only ATP/Ptotal was significant, moreover, if the Bonferroni correction for the number of tested parameters was applied, this parameter would not pass.
The reduced concentrations of PCr, ATP and Qmax indicate a pronounced limitation of metabolism in patients with a history/ongoing diabetic foot syndrome. The use of more accurate absolute quantification should improve the monitoring of potential treatment outcomes. An applicable method for the absolute quantification of phosphorus metabolites using an external standard was employed. When compared groups of subjects, the results revealed significantly reduced levels of PCr and ATP, and even Qmax in the cohort patients with a history/ongoing diabetic foot syndrome.
The study was supported by grants from the Ministry of Health, Czech Republic NW24-09-00184 and DRO („Institute for Clinical and Experimental Medicine – IKEM, IN 00023001“), Programme EXCELES - ID Project No. LX22NPO5104 - Funded by the European Union – Next Generation EU.
Petr SEDIVY (Prague, Czech Republic), Monika DEZORTOVA, Dita PAJUELO, Petr KORDAC, Vladimira FEJFAROVA, Taimr PAVEL, Milan HAJEK
14:06 - 14:51
Lightning talk Poster discussion.
|
Salle Major |
|
"Thursday 09 October"
Added to your list of favorites
Deleted from your list of favorites
D11
13:30 - 15:00
FT3 LT - Increasing quality when assessing microstructure
FT3: Cycle of Quality
13:30 - 13:32
#47619 - PG079 Enhancing Specificity in Fixel Based Analysis with Biophysical Modelling.
PG079 Enhancing Specificity in Fixel Based Analysis with Biophysical Modelling.
Fixel(fibre element contained within a voxel)-based analysis aims to extract information from voxelwise MRI data specific to fibre populations within a voxel. This has traditionally been done using constrained spherical deconvolution(CSD)[1]. We recently developed an alternative framework, WHIM[2], which fits a biophysical model to fixels using a hierarchical model to resolve fibre alignment across subjects. Despite the growing use of fixel-based analysis, the specificity of such analysis in isolating changes to affected fibres remains underexplored. A controlled mouse brain injury model[3] (Figure 1), in which one tract is selectively injured while the other remains intact, offers an ideal benchmark for assessing the sensitivity and specificity of fixel-based metrics. This setting allows us to evaluate and refine WHIM to improve fibre specificity, ensuring that detected changes are confined to the affected tract without cross-contamination from crossing fibres.
Data
We used publicly available white matter microscopy dataset [3, 4]. For this data unilateral retinal ischemia/reperfusion was induced in 10 rats by elevating the pressure in the anterior chamber of one eye for 90 minutes. Additionally, 5 rats served as controls. Five weeks post-injury, the rats were intracardially perfused for dMRI and histology measurements. This protocol yielded a total of 15 brain scans, each with 80 DWI directions at bvalues of 2000 and 2500 s/mm² and 20 b0 images, with an isotropic voxel resolution of 125 μm³.
Models
The basic model for fixel-based analysis in WHIM is the ball and sticks model [5] that includes a ball compartment, capturing isotropic diffusion, and a stick model for each fibre population with orientation and volume fraction as free parameters (Figure 2). The orientation parameters are estimated in a hierarchical framework to ensure correspondence in labelling across subjects/spatial location. All compartments share the same diffusivity.
Previous studies have suggested that Wallerian degeneration may increase perpendicular diffusivity by reducing axonal barriers [6]. Therefore, we incorporated a fixel-specific perpendicular diffusivity into the model. The updated model, therefore, includes voxel-wise parameters for isotropic and parallel diffusivity, along with fixel-specific parameters for volume fractions, perpendicular diffusivity, and orientation for each fixel as shown in Figure 2. A shrinkage prior was applied to constrain the perpendicular diffusivity estimates (truncated gaussian with mean=0 and std=0.1 μm²/ms).
In both models, orientations in the optic chiasm are estimated using an anatomically informed prior of 45 and -45 degrees in the axial plane with dispersion of 0.1. We first fitted the basic ball-and-stick model with 2 fibres to dMRI data from the optic chiasm, where the two optic tracts intersect. Then, the signal fractions for fixels oriented left-to-right and right-to-left were averaged in each brain. We found a significant reduction in the average signal fraction for the injured fibre population, confirming that this measure is sensitive to degeneration (Figure 3). However, the signal fraction for the intact fibre population increased, suggesting that this metric lacks fibre specificity and may be affected by changes in crossing fibre populations.
To improve specificity, we subsequently employed an extended ball-and-stick model, allowing each stick compartment to include perpendicular diffusivity as an additional free parameter. In this model the signal fraction decreased for the injured fibre population, while the signal fraction for the intact fibre remained consistent with control values. Furthermore, the model revealed an increase in perpendicular diffusivity in the injured fibre, supporting the hypothesis that Wallerian degeneration leads to increased water diffusion perpendicular to axons due to reduced myelination and axonal barriers. Our results show that signal fractions derived from conventional fixel models can exhibit cross-talk between fixels, due to model constraints such as the requirement that signal fractions sum to one. This limits the ability to assign changes specifically to affected fibres. By incorporating extra biophysical parameters; such as fixel-specific perpendicular diffusivity into the model; we reduce contamination from neighbouring fibres and gain metrics that are both sensitive and specific to underlying pathology. The observed increase in perpendicular diffusivity in the injured tract supports the hypothesis that Wallerian degeneration reduces axonal barriers, and suggests that these features can be captured by more flexible, biophysically-informed fixel models. Integrating biophysical modelling into fixel-based analysis improves its specificity in detecting tract-specific degeneration in crossing fibre regions. Our method, validated in a controlled injury model, provides a promising direction for developing more interpretable and specific diffusion MRI metrics.
Hossein RAFIPOOR, Michiel COTTAAR (Oxford, United Kingdom), Concha LUIS, Saad JBABDI
13:32 - 13:34
#46028 - PG080 Resolving partial-volume in the brain with multidimensional T1xT2 MRI.
PG080 Resolving partial-volume in the brain with multidimensional T1xT2 MRI.
The MRI contrast depends on T1 and T2 relaxation. Characterizing the relaxation processes is fundamental to understand the image contrast and variability.
T1 and T2 relaxation are often characterized with a single time constant1. However, many brain voxels contain several compartments2. These compartments are microscopic (intra/extra-celullar/myelin-water) and/or mesoscopic (white/gray matter, cerebrospinal fluid; WM/GM/CSF). This results in multiexponential behavior2,3.
Disentangling the microscopic contributions is an active research topic2,3. However, disentangling the mesoscopy (partial-volume) is also important: it may help model signal behaviour or probe the microscopy while accounting for mesoscopic contributions.
Deriving T1/T2 distributions from 1D decays (e.g. with inverse-Laplace fits) is inherently ill-posed2. In NMR, 2D measurements are instead commonly used. For relaxometry, this involves manipulating both inversion and echo time to derive a 2D-T1xT2 spectrum4. This better separates compartments5, since the multiplication of relaxation kernels increases the between-compartment signal difference. These approaches are making their way to MRI6–8.
Here we explore the ability of 2D-T1xT2 MRI to separate mesoscopic brain tissue compartments (WM/GM/CSF).
3 participants (1 male, age=33-36) were scanned (Siemens Prisma 3T, 32Rx head coil). The acquisition lasted ~1h15min.
For the 2D-T1xT2 measurements, an inversion prepared, spin-echo EPI (single-shot, matrix=110x110, GRAPPA=3, Partial-Fourier=6/8, single-slice, slice-thickness=4mm, voxel-size=2x2x4mm3, TR=13sec, bandwidth=2392Hz/Px, nonselective adiabatic inversion) was repeated for combinations of logarithmically-spaced inversion times (N=9, TI=[36-3000]ms) and echo times (N=10, TE=[15-300]ms; Fig.1A-B). The single-slice excitation/refocusing avoided between-slice crosstalk. The slice was placed 2mm above the AC-PC line. The long TR ensured close-to-complete brain-tissue relaxation. No fat suppression was employed to avoid magnetization transfer effects.
1D-T1 and 1D-T2 measurements were performed with the same protocol (1D-T1: 26 TIs=[21-3000]ms and TEs=[15-300]ms).
The IR-prepared data were PSIR-reconstructed. For the 2D-T1xT2, 1D-T1 and 1D-T2, a kernel describing the signal as a distribution of time constants was fitted (NNLS; quadprog in R; Fig.1C). Solution-smoothness was imposed with L2. For comparison, the standard single-exponential model was also fitted for 1D-T1/1D-T2.
To validate our fitting, we combined signal from WM, GM and CSF ROIs to simulate multicompartment data.
We then averaged the spectra across voxels and extracted 5 components (k-means; component number empirically determined). These spectral components were matched to WM, GM and CSF based on relaxometry values from IR/SE-EPI literature review9 (T1=650-950ms/T2=45-75ms; T1=1000-1500ms/T2=55-120ms; T1>1500ms/T2>100ms respectively). Multi-exponential fits reduced residuals compared to single-exponential across ROIs (Fig.2A-C). The 2D spectrum resolved 3 peaks (peaks: T1/T2=847/54ms; T1/T2=1215/53ms; T1/T2>2000/200 matching the literature values9 albeit with shorter T2GM, Fig.2E). 1D-T1 and 1D-T2 spectra similarly produced peaks matching the literature, but could separate only 2 peaks.
In anatomical-border voxels (WM/GM; GM/CSF), 2D-T1xT2 spectra consistently showed multiple peaks (Fig.3). 1D-T1 spectra did not reliably differentiate WM/GM or GM/CSF peaks. 1D-T2 spectra often produced an extreme peak likely due to fitting-range bias.
K-means clusters from 1D-T1, 1D-T2 and 2D-T1xT2 were consistent across individuals. The clusters overlapped with WM, GM and CSF, confirming fit sensitivity to anatomical properties. In 2D-T1xT2, the iron-rich basal ganglia formed a separate cluster in two individuals (black arrows), while a fifth cluster likely represented fitting range biases. Differentiating the basal ganglia in 1D-T1/1D-T2 was unfeasible. Reliable WM/GM separation with 1D-T2 was not possible. Peak cluster amplitudes decreased near anatomical borders, suggesting partial volume (Fig.4B).
The spatial map of voxels with peaks in both CSF and GM components or WM and GM, closely tracked anatomical boundaries (i.e. CSF/GM boundary-red; WM/GM-blue; Fig.4C). This suggests that 2D-T1xT2 spectra disentangle partial volume within-voxel. The WM/GM boundary was less clear, likely due to similar relaxometry properties. In brain MRI, voxels commonly include multiple mesoscopic compartments due to resolution limits. Our results suggest that 2D-T1xT2 can leverage the increased signal specificity to relaxometry properties (compared to 1D-T1/1D-T2) to resolve these tissue types within-voxel. This may be crucial to understand and model signal behaviour.
Further expanding 2D-T1xT2 to shorter TIs/TEs may increase sensitivity to microscopic compartments (e.g. myelin2,3). This can also provide a ground-truth testbed, where microscopic compartments can be probed while ensuring the mesoscopy is the same.
Nikos PRIOVOULOS (OXFORD, United Kingdom), Hanna LIU, Dan BENJAMINI, Amy HOWARD, Karla MILLER, Aaron HESS
13:34 - 13:36
#47646 - PG081 Evaluation of different partial volume correction methods for sodium MRI using a resolution phantom.
PG081 Evaluation of different partial volume correction methods for sodium MRI using a resolution phantom.
Sodium MRI is a functional imaging technique that can provide physiological information. Due to low inherent signal and short sodium relaxation times, large voxel sizes and radial acquisition techniques are widely used [1]. This acquisition leads to substantial partial volume effects (PVE), which are particularly problematic in small structures such as cartilage or tendons [2]. The large voxel sizes lead to intra-voxel tissue averaging, the so-called “tissue-fraction” effect, while radial acquisitions have an inherently broader point spread function (PSF) leading to signal spilling (“spill-over” effect) [3]. In recent years several partial volume correction (PVC) methods have been developed that focus on correcting these effects with varying approaches. The purpose of this study was to evaluate four of these using a resolution phantom.
A resolution phantom filled with sodium chloride solution was imaged using a 23Na/1H-surface coil (RAPID Biomedical GmbH) and a density-adapted 3D radial sequence [4] at a 3 T MRI (Siemens MAGNETON Prisma) at 2 x 2 x 2 mm3 resolution. Proton images for segmentation were acquired at 1 x 1 x 1 mm3 resolution. The phantom consisted of 1 to 6 mm wide lamellae with the same spacing as width (Figure 1). PVC methods were a geometric transfer matrix (GTM) [5], a modulated least trimmed square (3D-mLTS) linear regression [3], a region correction based on the proton-sodium segmentation ratio (PSSR) [2] and a combination of the first two (mLTS-GTM). The GTM method corrects “spill-over” artifacts by convolving a simulated PSF with a mask of the image region. Based on this, a set of linear equations can be constructed from the spill contributions of each compartment that form the GTM. By matrix inversion the original signal can be recovered [5]. The other methods consider the tissue fraction effect, where the mLTS is a pixel-wise correction that recovers the signal based on a 3 x 3 x 3 voxel kernel using linear regression resulting in a PVE-corrected image [3]. The PSSR calculates a weighting factor based on the volume ratio of the higher resolution mask of the proton image to the lower resolution of the sodium image [2]. All methods were applied to the phantom MR images and evaluated with a modulation transfer function (MTF) [6]. Apparent sodium concentrations (ASCs) were calculated. The modulation transfer function shows a clear decrease in contrast between high signal and low to zero signal regions without PVC (Figure 2). Note that the 1 mm lamella was not included, because it is not visible in the proton image and therefore cannot be segmented. With the exception of the mLTS method, all correction methods result in a smaller decrease in the MTF and therefore a greater transfer of contrast. The ASC is diminished relative to the expected value of 154 mM without PVC, particularly within the slimmer lamellae (Table 1, Figure 2, Figure 3). All PVC methods increase the concentration of all lamellae, with the GTM showing the greatest improvement and the mLTS showing only a very slight improvement of 1-5 mM compared to no correction. However, none of the methods were able to fully restore the signal intensity to its expected value, there was no overestimation. The maximum value achieved was 149.8 mM using the GTM method in the 5 mm lamella. All methods improve the transfer of contrast as well as the accuracy of ASC determination. The GTM is the most widely used method, but has the disadvantage of not providing a PVE-corrected image, which is possible with the mLTS [3]. However, it has been developed on cartesian images with a narrower PSF. The GTM method shows the greatest improvement in both respects but cannot fully recover the signal in lamellae with a width in the same order of magnitude as the image resolution, such as the 2 mm lamellae (Figure 2), where the ASC is only 50 % of the nominal value. Both PSSR and mLTS target the “tissue fraction” effect and achieve less improvement than the GTM. This suggests that the “spill-over” effect is dominant in this type of imaging. This effect is likely to be the reason for the low impact of the mLTS, as it takes the signal in a small surrounding kernel into account. Significant signal spill-over due to the PSF leads to similar values inside and outside the segmentation in small regions so that the regression has little effect. As the PSSR inherently assumes zero surrounding signal, it is not affected by the spill-over. The combination of mLTS and GTM gives similar results to the GTM alone, which is to be expected as the mLTS has little effect on itself. PVC can generally improve the transfer of contrast and ASC determination in sodium imaging. Correcting “spill-over” artifacts has a greater impact, because spill-over of the signal with the point spread function is the dominant PVE in radial imaging. The GTM method gives the best results and is therefore recommended for correction.
Rika MÖLLER (Düsseldorf, Germany), Paula LEJA, Benedikt KAMP, Mohammed Ali GOUNDI, Armin NAGEL, Hans-Jörg WITTSACK, Anja MÜLLER-LUTZ
13:36 - 13:38
#47914 - PG082 Quantitative MRI of the human brain ex vivo and in vivo: the role of local magnetic field gradients.
PG082 Quantitative MRI of the human brain ex vivo and in vivo: the role of local magnetic field gradients.
MRI of human ex vivo tissue can be of value for pushing MRI resolution limits and for validation based on histology [1,2] and µCT3 of the same specimens. MRI at 9.4T in vivo is compatible with whole brain ex vivo MRI, revealing absence of significant variation in cortical thickness and maintained QSM sensitivity to expected iron concentrations [4]. To further our understanding of these observations, we extended the previous comparison to include R2 and R2’ mappings and qMRI at 3T, where the contributions from different relaxation mechanisms are different than at 9.4T.
Ex vivo whole brains (N=4, 80±1y 2F, PMI≤12h) were immersion fixed in formalin with known dielectric properties [5]. Five young (31±8y, 2F) and five elderly (69±4, 2F) healthy subjects volunteered to participate in agreement with Ethics approval from the Medical Faculty, University of Tübingen.
Quantitative maps, R2*, QSM, R2 (using the EPG algorithm) [6,7] and R2prim were obtained at 3T and 9.4T (MRI protocols in Table 1).
Tissue segmentations were obtained from skull-stripped [8] MP2RAGE contrast and R1 images with CAT12 (version 12.9, in Matlab R2018b), LN2_RIM_POLISH [9] and resliced into 3D and 2D space (trilinear interpolation). Inverse spatial deformation fields were used to obtain brain parcellations in native space in 3D and 2D space. qMRI data were extracted from tissue regions, and age-based tissue iron concentrations were generated [7,10] R2 relaxation, which is caused by adiabatic exchange and tumbling at the Larmor frequency, remained stable during fixation with in vivo-like values (Fig 1a). After >65 days of fixation, also R2* and R2prim stabilized, albeit at values greater than in vivo. The largest relative difference compared to in vivo occurred for R2prim in the white matter (Fig 1b). Both R2 and R2prim values were higher in brain regions with higher age-based estimates of the iron concentration (Fig 2). Ex vivo, a large variability in relaxation rates was observed overall. With respect to R2, the sensitivity of R2prim to iron was greater at 9.4T than at 3T, with a clear R2prim dominance over R2 in subcortical regions. Compared to the measured magnetic susceptibility ex vivo and in vivo, the R2* was smaller than what is expected for the static dephasing regime [11]. For instance, in the globus pallidus a very small dephasing effect was observed despite its high iron concentration (Fig 3). Quantitative MRI at different magnetic field strengths offer unique capabilities for studying tissue properties. Although ex vivo MRI of formalin fixed whole brain specimens offers a series of advantages, like high resolution scans for comparison with histology, may not allow to maintain all magnetic properties unchanged. In this respect, R2 which is deployed at the molecular level and reflects relaxation caused by molecular tumbling at the Larmor frequency (and twice its value) and adiabatic processes, was found to be preserved ex vivo. R2*and Rprim that are sensitive to local magnetic field gradients underwent dynamic changes during immersion fixation with an initial increase, followed by decrease before tend to stabilize after ca 65 days. At the cortical surface, the relationship between R2* and QSM was close to values expected in the static dephasing regime, while values compatible with the presence of dynamic averaging effects were observed in the globus pallidus. Since this structure is located far away from brain surfaces through which the fixative enters the tissue, this observation could possibly indicate dynamic averaging processes in presence of tissue alterations like cell swelling. Ex vivo MRI with formalin fixed whole brain specimens can be of value for validation purposes, and can give indications about tissue status. Further studies are warranted to advance our knowledge about how transverse relaxation mechanisms unfold in vivo and ex vivo.
References
1. Bagnato F, et al. Brain, 2011;134(12):3602-3615. doi:10.1093/brain/awr278 ;
2. Hametner S, et al.. Neuroimage. 2018;179(May):117-133. doi:10.1016/j.neuroimage.2018.06.007 ;
3. Lee JY, et al.,Front Neurosci. 2023;17. doi:10.3389/fnins.2023.1236876 ;
4. Hagberg GE, et al., In: Magnetic Resonance Materials in Physics, Biology and Medicine. Vol 37. ; 2024:136-137. doi:10.1007/s10334-024-01191-6 ;
5. Nazemorroaya A, et al., Magn Reson Med. 2022;87(5):2481-2494. doi:10.1002/mrm.29122 ;
6. Hagberg GE, et al., Neuroimage. 2017;144:203-216. doi:10.1016/j.neuroimage.2016.09.047 ;
7. Hagberg GE, et al., Magn Reson Med. 2022;88(5):2267-2276. doi:10.1002/mrm.29368 ;
8. Hoopes A, et al., Neuroimage. 2022;260:119474. doi:10.1016/j.neuroimage.2022.119474 ;
9. Huber L et al.,. Neuroimage. 2021;237:118091. doi:10.1016/j.neuroimage.2021.118091 ;
10. Hallgren B, Sourander P. J Neurochem. 1958;3(1):41-51. doi:10.1111/j.1471-4159.1958.tb12607.x ;
11. Duyn JH, Schenck J. NMR Biomed. 2017;30(4):e3546. doi:10.1002/nbm.3546
Gisela E HAGBERG (Tübingen, Germany), Shiozawa THOMAS, Bernhard HIRT, Klaus SCHEFFLER
13:38 - 13:40
#45710 - PG083 IQT for Diffusion MRI: The Impact of Physics-Based Augmentation and Batch Optimisation.
PG083 IQT for Diffusion MRI: The Impact of Physics-Based Augmentation and Batch Optimisation.
Deep Learning approaches for post-reconstruction enhancement of diffusion MRI (dMRI) have shown promising results in the past, with the possibility of enhancing contrast, information content, spatial resolution and/or accelerating dMRI acquisition. Particularly Image Quality Transfer (IQT) [1, 2] enhances the resolution and contrast of images through a computational model often learned from artificially downsampled data. Recent work [3] shows that IQT for dMRI can be improved by incorporating high resolution structural MRI as a secondary input to guide the resolution and contrast enhancement.
However, prior IQT implementations and super-resolution/enhancement approaches in general for dMRI [4] have learned to upsample low-resolution images by a single upsampling factor and worked on model parameters (typically diffusion tensor components) rather than raw dMRI data, due to smaller number of parameters and independence from an acquisition protocol. Previous work [3] shows benefits of the model learning to upsample using multiple factors in certain configurations, but the choice of representation space (tensor components versus raw dMRI) and their effect on prediction performance have not been investigated.
Here we evaluate different augmentation approaches for training the multimodal IQT framework [3]. We test various configurations to determine the optimal training strategy for realistic enhancement of dMRI.
The training data comes from the Human Connectome Project [5] young adult cohort. We use dMRI and T1-weighted images from a total of 159 subjects, with a training, testing, and validation split of 70%, 20%, and 10% respectively. We evaluate the performance of the data representation strategy on multimodal IQT [3] without the addition of noise augmentation to input data by upsampling from low resolution images with isotropic voxel spacing of 2.5mm to 1.25mm for testing.
We compare the performance of the IQT model on 3 different training configurations as independent variables:
1) The augmentation space. We compare the performance of applying data preprocessing methods on the raw dMRI data prior to model fitting, which we refer to as q-space, vs the fitted Diffusion Tensor Imaging [6] tensor parameters (tensor space).
2) The number of learned upsampling factors per batch. We compare the performance of our model trained using the same upsampling factor per batch vs trained using different upsampling factors per image within a batch.
3) The acquisition strategy for the upsampling factors. We compare the model performance with downsampling rates sampled from a set of discrete values and with downsampling rates sampled from a continuous uniform distribution.
Our baseline approach is the model that uses q-space sampled data, with different upsampling rates for each image within a batch, acquired from the set of 4 different upsampling rates from the multimodal IQT framework [3].
We evaluate the model’s performance on raw data using our DT-RMSE metric [1], and on processed metrics using the structural similarity index measure [7] for mean diffusivity (MD), fractional anisotropy (FA) and cosine similarity for coloured FA (CFA) [3, 8]. We evaluate model performances using the median values of the testing dataset per each metric. Quantitative results from Figure 1 indicate that the best performance for DT-RMSE is achieved by the model trained on data sampled in q-space, with multiple learned upsampling rates within each training batch, where the upsampling rates were sampled from a continuous uniform distribution.
Qualitative results from Figures 2 and 3 indicate that certain structures such as the cerebellar tentorium is captured better with q-space sampling in contrast to tensor space sampling, where said structure does not exist. However, qualitative results do not demonstrate a noteworthy difference between using set of values and a continuous distribution. IQT achieves better performance when the training data is sampled in q-space compared to sampling in tensor space, indicating that the IQT model captures DTI data better during training on fitted dMRI data sampled in q-space.
IQT achieves better performance when it learns a different upsampling rate with each image compared to learning a single upsampling rate per batch. Compared to using multiple upsampling rates per batch, changing the acquisition strategy for upsampling rates has a smaller effect on results. This work explores dMRI sampling strategies for IQT. Results show that sampling in q-space is optimal for preserving diffusivity values , and that incorporating multiple upsampling rates within each training batch leads to a notable improvement in image quality. Future work includes testing on datasets the network is not familiar with, the incorporation of augmentation to training data such as random noise injection to mimic the acquisition of more realistic dMRI data, and experimentation on multi-shell approaches such as MAP or SANDI [9, 10].
Alp G. CICIMEN (London, United Kingdom), Henry F. J. TREGIDGO, Matteo FIGINI, Marco PALOMBO, Derek K. JONES, Daniel C. ALEXANDER
13:40 - 13:42
#46905 - PG084 Diffusion tensor imaging of the spinal cord with a no-b-zero approach can reduce the impact of CSF pulsations and partial volume effects.
PG084 Diffusion tensor imaging of the spinal cord with a no-b-zero approach can reduce the impact of CSF pulsations and partial volume effects.
Diffusion tensor imaging (DTI) of the spinal cord is highly susceptible to partial volume effects (PVE) due to often limited spatial resolution and the distinct diffusion properties of the surrounding cerebrospinal fluid (CSF). In addition, the pulsations of the CSF flow induce spatial variation in the diffusion parameters, mean diffusivity (MD), and fractional anisotropy (FA). The pulsations also limit the efficacy of postprocessing corrections, such as the free water elimination model[1].
For brain DTI, it has been shown that a no-b-zero (NBZ) approach, where the signal at b = 0 s/mm2 (b0) is excluded, can effectively reduce the influence of PVE in MD estimations [2], [3]. It is plausible that this method could reduce PVE in spinal cord DTI and reduce the impact of CSF pulsations in DTI estimations of the spinal cord, especially when gating or cardiac triggering is not used.
This study aims to determine whether the choice of b-values in DTI evaluations of the spinal cord has the feasibility to reduce both PVE and bias from the CSF pulsations in MD and FA measurements.
Simulations of PVE were performed using a bi-exponential model with different combinations of b-values (0, 200, and 800 s/mm2), number of gradient directions (6 and 32), and varying fractions of CSF and spinal cord signal.
Two healthy volunteers were scanned in a Philips MR-7700 using a multi-shot EPI with a reduced field-of-view sequence (Figure 1), a research software patch enabled a multi-shell acquisition with custom diffusion directions. To investigate the effect of excluding b0, MD and FA were measured in regions of interest (ROIs) drawn in the CSF, spinal cord, and at the border between them, where PVE is expected to be prominent (Figure 1). All DTI parameter estimations were performed with the DIPY toolbox [4]. Paired t-tests were used to evaluate differences in the ROI measurements between including and excluding b0. A p-value <0.05 was considered statistically significant. The simulations showed that the choice of b-value and the number of gradient directions had an impact on the observed MD in voxels with PVE (Figure 2). While combinations of all three b-values displayed lower MD values than b0-200 and b0-800, the largest reduction was achieved when b0 was excluded from the DTI fit, especially when fewer gradient directions were used (Figure 2).
For both volunteers and all ROIs, MD was higher when b0 was included compared to excluded in the DTI fit (Figure 3). The changes were largest in the CSF and PVE ROIs, but significant in all ROIs. The difference in FA values when excluding b0 was minimal and not significant, except for in the PVE ROI (Figure 3).
There are visible changes in the MD map if b0 is excluded from the DTI fit (Figure 4A). The largest differences in the MD maps were observed in the CSF close to the discs. Visible changes in the FA maps when excluding b0 were comparably very small (Figure 4B). The results of the simulation indicate that MD values in voxels with PVE between spinal cord and CSF are influenced by the choice of b-value and number of diffusion directions (Figure 2). Suggesting that using a multi-shell diffusion scheme is beneficial to reduce MD values, even more so if using the no-b-zero approach, agreeing with previous studies [2], [3].
The in vivo results confirmed the simulations by showing that exclusion of b0 leads to a significant decrease in MD values (Figure 3) and the largest differences was found in the borders between CSF and spinal cord or other tissue as well as in CSF near discs (Figure 4), consistent with where PVE are expected to be largest. Exclusion of b0 also reduced some of the visible MD heterogeneity (Figure 4) in the CSF, suggesting that this approach also reduces the impact of CSF pulsations in the DTI estimations. This is particularly important in patients with spinal pathology, where CSF pulsations may be affected by the pathology, introducing additional bias in the MD estimates via the PVE. The results indicate that the non-b-zero approach can be used to mitigate pulsations when cardiac triggering is not used.
FA values were substantially less affected by the b-value selection and the exclusion of b0, indicating that a different approach might be necessary to reduce the impact of PVE and CSF pulsations in FA maps. The preliminary results of this study suggest that using a no-b-zero approach and excluding b0 from the DTI-fit is a very promising, effective, and simple way to reduce the impact of PVE, and most importantly, the impact CSF pulsations on the MD maps of the spinal cord.
Alma BLOMBÄCK (Gothenburg, Sweden), Maria LJUNGBERG, Kerstin LAGERSTRAND
13:42 - 13:44
#47817 - PG085 Denoising for High-Resolution Tensor-Valued Diffusion on Medium Gradient MRI Scanners.
PG085 Denoising for High-Resolution Tensor-Valued Diffusion on Medium Gradient MRI Scanners.
Tensor-valued diffusion MRI (dMRI) enables detailed characterization of brain tissue microstructure but faces challenges in achieving high spatial resolution due to inherently low signal-to-noise ratio (SNR). This limitation is particularly pronounced on MRI systems with gradient performance around 40 mT/m, often used in radiotherapy settings. We aimed to evaluate the effectiveness of denoising methods—including PCA-based and deep-learning techniques—to enhance the resolution of tensor-valued diffusion MRI on medium-gradient scanners.
We assessed four denoising methods: Marchenko-Pastur PCA (MPPCA) from MRtrix3 [1], MPPCA from the NeuroPhysics group at Aarhus University (MPPCA_CFIN) [2], Noise Reduction with Distribution Corrected PCA (NORDIC) [3], and a commercial deep learning-based method (AirReconDL, ARDL, GE Healthcare, Milwaukee, WI, USA) [4]. ARDL denoising was inherently applied to complex data during the MRI scanner reconstruction process, and NORDIC was exclusively applied to complex data post-reconstruction. MPPCA and MPPCA_CFIN were applied to both magnitude and complex images post-reconstruction.
Data were collected on a 3T Signa Architect MRI scanner (GE Healthcare, Milwaukee, WI, USA) using both diagnostic and radiotherapy coil setups, with resolutions from 3x3x3 mm³ down to 1.8x1.8x1.8 mm³. Phantom measurements evaluated denoising impact on the noise floor and parameter biases, while in vivo brain imaging assessed denoising effectiveness in clinical conditions. Tensor-valued diffusion parameters Mean Diffusivity (MD), Fractional Anisotropy (FA), microscopic Fractional Anisotropy (µFA), isotropic Mean Kurtosis (MKI), and anisotropic Mean Kurtosis (MKA) were analyzed [5].
The MRI protocol employed linear tensor encoding (LTE) and spherical tensor encoding (STE) sequences. Data were acquired at multiple b-values (100, 700, 1400, and 2000 s/mm²), collected in 6, 6, 10, and 21 directions for LTE, and 6, 6, 10, and 15 unique rotations for STE. Additionally, spherical diffusion encoding at b=2000 s/mm² was specifically acquired to evaluate noise characteristics. The SNR was systematically modulated by adjusting the transmit gain during acquisitions, allowing investigation of the effect of decreased gain on image quality and parameter accuracy.
Signal precision metrics were defined as Q3 and Q6. Q3 represented the fraction of voxels with an SNR greater than 3, indicating regions where the noise distribution could reasonably be approximated as Gaussian. Q6 represented the fraction of voxels with an SNR greater than 6, serving as a more stringent measure to ensure robust parameter estimation. Complex-valued denoising methods consistently outperformed magnitude-based methods in reducing noise floor effects and parameter estimation bias (Figure 1). Phantom studies revealed that magnitude denoising reduced ADC variance but did not overcome noise-floor bias, whereas complex-valued methods effectively mitigated these biases (Figure 2). Signal precision metrics (Q3 and Q6) showed marked improvements across all resolutions with MPPCA_CFIN denoising, maintaining high voxel fractions with acceptable SNR (Figure 3). Parameter-specific analyses indicated robust performance of MD and µFA across methods; however, FA demonstrated significant method-dependent variability. MKI showed the greatest sensitivity to noise and limited improvements from denoising methods. Visual assessment of QTI parameter maps confirmed substantial improvements in detail and contrast with complex-valued denoising, particularly evident at higher resolutions. MPPCA_CFIN demonstrated particularly strong performance, enabling high-quality parameter estimation at 2x2x2 mm³ resolution using radiotherapy coil setups and at 1.8x1.8x1.8 mm³ with diagnostic setups, comparable to conventional 3x3x3 mm³ acquisitions (Figure 4). Our findings highlight the substantial advantages of complex-valued denoising methods over magnitude-based approaches for high-resolution tensor-valued diffusion MRI. Complex-valued methods effectively address noise-floor issues, critical for accurate diffusion parameter estimation, especially at high resolutions and b-values. Despite these improvements, MKI remain challenging due to intrinsic noise sensitivity. The MPPCA_CFIN method showed superior consistency across resolutions and setups, suggesting it as an optimal solution for clinical adoption. The capability to achieve higher resolutions without compromising parameter accuracy offers significant potential for improving tissue characterization and clinical decision-making in radiotherapy. Complex-valued denoising improves the achievable spatial resolution of tensor-valued diffusion MRI on medium-gradient MRI systems, enhancing its utility in treatment planning and early response monitoring in radiotherapy. Despite notable improvements, parameter-specific limitations persist, especially regarding MKI estimation.
Patrik BRYNOLFSSON (Lund, Sweden), Ivan RASHID, Minna LERNER, Lars OLSSON
13:44 - 13:46
#47742 - PG086 Investigating orientation dependency in X-separation for myelin quantification in brain white matter.
PG086 Investigating orientation dependency in X-separation for myelin quantification in brain white matter.
Susceptibility source separation (X-separation) is a recent advancement of Quantitative Susceptibility Mapping (QSM). While QSM assesses the bulk magnetic susceptibility, a composition of paramagnetic (Xpara) and diamagnetic (Xdia) contributions, X-separation separates these components by integrating the local magnetic field and transverse relaxation rates (R2* or R2’) [1,2]. In the human brain, magnetic susceptibility is primarily linked to two key tissue components: iron, which exerts paramagnetic effects, and myelin, which has diamagnetic properties. While QSM assesses the tissues bulk magnetic susceptibility, a composition of paramagnetic and diamagnetic contributions, X-separation allows a separation of these components. This capability enables specific biomarker quantification, such as Xdia as a proxy for myelin content [3], addressing a critical limitation of QSM or R2* mapping in resolving counteracting contributions of iron and myelin [4].
In white matter (WM), myelin’s diamagnetic properties make Xdia a promising non-invasive marker for demyelinating diseases such as Multiple Sclerosis [5]. However, the anisotropic microstructure of WM introduces well documented orientation-dependent effects in quantitative MRI parameters, including frequency shifts (Δfz) and R2* [6,7]. While Δfz and R2* exhibit orientation dependence, it remains unclear whether X-separation inherit or mitigate orientation effects. Thus, the purpose of this study was to investigate the orientation dependency of Xpara and Xdia, assessed with different X-separation algorithms, in WM.
Seven healthy volunteers (3 female, 4 male, age 24–61) were scanned on a 3T MRI system (MAGNETOM Skyra, Siemens). Diffusion tensor imaging (DTI) was acquired (b = 1000 s/mm², 30 directions, TE = 92 ms, TR = 9600 ms, 2 mm isotropic) to assess fiber orientation. A T1-weighted MPRAGE (TE = 2.12 ms, TR = 1690 ms, flip angle = 8°, 0.8 mm isotropic) was used for WM segmentation. For X-Separation, a 3D multi-echo GRE (6 echoes, TEs = 4.92–29.51 ms, TR = 35 ms, 1 mm isotropic) and a dual-echo spin-echo sequence (TE = 11/100 ms, TR = 5890 ms, 1 × 1 × 2 mm³) were acquired to compute R2*, QSM, Δfz, and R2. R2 was estimated via dual-echo fitting; R2* via the ARLO algorithm [8].
We compared four X-separation approaches: SepNet-R2*, SepNet-R2′ (both neural networks), the original Chi-separation with MEDI, and an iLSQR-based method. Maps were generated using the ChiSep Toolbox[1]. DTI data were denoised and corrected; fiber orientation was calculated as the angle between the main magnetic field and the first eigenvector. WM segmentation used FSL FAST with FA > 0.5. R2* and R2′ exhibited clear orientation dependence, following a sin⁴(θ) pattern, while Δfz showed a sin²(θ) trend, consistent with WM anisotropy models (Figure 1). Figure 2 presents the angular behavior of Xpara, Xdia, and Xtotal across four X-separation algorithms. Despite differences in absolute values, Xpara consistently showed a sin⁴(θ) dependency, mirroring R2* and R2′, while Xdia followed a mixed sin²(θ) and sin⁴(θ) pattern, reflecting myelin’s anisotropy. Xtotal varied less with angle, though neural network models (e.g., SepNet) peaked at lower orientations than iterative methods (e.g., iLSQR), indicating differences in sensitivity to orientation effects. This study demonstrates that both Xpara and Xdia show orientation dependence in WM. Surprisingly, Xpara—which reflects iron and is assumed isotropic—exhibits a clear sin⁴(θ) dependency, mirroring R2* and R2′ trends. This challenges the assumption of isotropic paramagnetic sources and likely stems from X-separation models neglecting microstructural anisotropy. Myelinated axons induce anisotropic magnetic environments that influence Δfz and relaxation, and although iron is isotropic, its alignment with myelinated structures may bias Xpara estimates. Most models, based on static dephasing theory, overlook these effects, leading to systematic errors in Xpara. In contrast, Xdia follows a mixed sin²(θ) and sin⁴(θ) dependency, consistent with theoretical expectations [6]. Our findings challenge the paradigm of orientation-independent paramagnetic susceptibility in X-separation, revealing that unmodeled microstructural anisotropy introduces significant orientation-dependent biases in Xpara and might underestimate the anisotropy in Xdia. To further enhance X-separation, future algorithms must account for anisotropy effects in WM. Addressing these limitations will enhance the specificity of Xdia as a myelin biomarker and improve the accuracy of iron quantification in disorders like MS, where disentangling these components especially in WM is clinically vital.
Alexander STÜRZ (Innsbruck, Austria), Bauer MELANIE, Christoph BIRKL
13:46 - 13:48
#47835 - PG087 Improving structural connectivity analysis in patients with mild cognitive impairment and dementia using multi-level thresholding and subcortical white matter segmentation.
PG087 Improving structural connectivity analysis in patients with mild cognitive impairment and dementia using multi-level thresholding and subcortical white matter segmentation.
Whole brain structural connectivity analysis using diffusion Magnetic Resonance Imaging (dMRI) tractography is a promising avenue of research, but several confounding factors limit the generalization of findings [1]. Potential false positive and false negative connections even with advanced tractography methods can lead to questionable and controversial results. A general approach to mitigate this uncertainty is employing group-wise thresholding but this could potentially lose information about weaker connections or subtle changes in select brain areas.
The selection of the brain parcellation scheme for defining nodes of the structural connectome can greatly influence results [2, 3]. Template-based atlas registration (like AAL3 [4]) is a simple and robust approach, but may lead to mislabeling of cortical and subcortical structures. Surface-based segmentation could be more anatomically correct describing individual brain structure, e.g. in neurodegenerative diseases, but labeling voxels based on tissue class yields regions of interest (ROIs) that only cover the gray matter – voxels that reconstructed fiber tracts potentially does not reach due to low anisotropy values in subcortical white matter.
In our study, the effect of adding subcortical white matter voxels to cortical ROIs on false negative connections was examined by creating six different parcellations. After whole brain CSD-tractography, resulting brain graphs were analyzed in a multi-threshold scheme, assessing inter-subject variability and false positive connections, in a cohort with healthy controls and patients with amnestic or non-amnestic type mild cognitive impairment (aMCI, naMCI), and dementia (DEM).
Data from 76 elderly subjects (mean age 68.4, 43 males, 25 controls, 18 naMCI, 22 aMCI, and 11 DEM, categorized by Addenbrooke Cognitive Examination) were collected using the Philips Ingenia 3T scanner at the Medical Imaging Centre of Semmelweis University. dMRI acquisition included 64 directions with b=2000s/〖mm〗^2 and 11 interspersed b=0 images, with 2mm isotropic resolution in 70 axial slices. T1 weighted 3D gradient echo images were also collected with 1mm isotropic resolution.
Diffusion data processing was performed in ExploreDTI [6, 7], including motion and distortion correction, using T1w images as registration targets. Whole-brain CSD tractography was performed, followed by calculation of connectivity matrices. 84 ROIs were defined using the Desikan-Killiany atlas [5] from Freesurfer segmentation [8] of T1w images. ROIs covering the cortical GM were amended by assigning voxels from the neighboring subcortical WM based on Euclidean distance in five, 1mm steps (Figure 1) using in-house Matlab scripts.
Multi-level thresholding of the connectivity matrices was performed based on connection density (number of interconnecting tracts / ROI surface) at 30 values equally spaced on logarithmic scale, followed by the calculation of graph theory metrics with Brain Connectivity Toolbox [9] and their analysis using non-parametric statistical tests between subject groups and ROI sets. Multi-level thresholding yielded three ranges of different network behavior consistent in each subject group with all six parcellations. With the most strict threshold values, only sparse connections were observed; between 4.55-3.79E-6 and 7.35E-7 threshold levels, the number of nodes in the network increased exponentially; finally, with lenient thresholds, only subtle differences were identified. The highest number of nodes increased with the inclusion of subcortical voxels (largest median network size: 77, 80, 82, 82, 83 and 84, respectively) (Figure 2).
Average characteristic path length decreased with added subcortical voxels and lenient thresholds. Kruskall-Wallis tests with post-hoc comparisons using Bonferroni-correction demonstrated that the most significant differences (p=0.0016) between HC and aMCI groups were observed with 2-3mm ROI amendment and moderate tract density thresholds (Figure 3), while the strongest difference (p=0.0007) between HC and DEM groups was without ROI amendment. Adding even a 1 mm wide sheet of subcortical WM voxels to GM ROIs limited the occurrence of nodes missing from the graphs by a factor of 5-10 (from around 11 subjects to 1 or 2) , suggesting that it lessens individual variability to at least some degree. Importantly, adding WM voxels had no effect on core network structure. However, above 3mm the amount of possible mislabeling and false connections may outweigh any benefits. Multi-level thresholding proved efficient in probing network structure across different connection strengths aiding the differentiation between subject groups. The combination of ROI amendment by subcortical WM segmentation and multi-level thresholding substantially decreased between subject variability, strengthening the results of between group comparisons in the graph theory-based analysis of patients with mild cognitive impairment and dementia.
Gyula GYEBNÁR (Budapest, Hungary), Noémi GYÜRE, Lajos Rudolf KOZÁK, Gábor CSUKLY
13:48 - 13:50
#47786 - PG088 Generalizable, Cross-sequence Physics-Informed Quantitative MRI Super-resolution.
PG088 Generalizable, Cross-sequence Physics-Informed Quantitative MRI Super-resolution.
Quantitative MRI maps (qMRI) captures intrinsic tissue properties (e.g., PD, T1, T2) enabling cross-sequence and cross-scanner comparison [1]. However, qMRI is often acquired at low resolution to limit scan time, while weighted images (T1w, T2w) are acquired at higher resolution and provide structural detail. Existing super-resolution methods for qMRI rely on high-resolution ground truth maps [2], which are difficult to obtain, particularly from patients. We introduce PHIRE-Q (PHysics-Informed super-Resolution for qMRI), which uses high-resolution weighted images to guide qMRI super-resolution in a self-supervised manner, eliminating the need for high-resolution qMRI. We train and validate PHIRE-Q on synthetic 2D qMRI (QRAPMASTER) [3] and test its generalizability with a MUPA 3D qMRI acquisition [4].
After model validation using image quality metrics (SSIM [7], HFEN [8]), its generalizability was assessed on qMRI acquired with MUPA and high-resolution T1w and T2w guide images, acquired as W. We compare the output (Q_h ) ̂ with a higher-sampled version of the MUPA maps (reference, Q_h). To further assess whether high-frequency anatomical details from the guide images were captured, weighted images were synthesized from Q_h, (Q_h ) ̂, Q_l using the signal model S with the acquisition settings of the guide images. These synthesized images were then visually compared with the actual guide images.
qMRI Qh (=PD, T1, T2) from 27 volunteers (10 for training, 17 for validation; 726 slices) were acquired using the QRAPMASTER sequence. T1-weighted (TR = [360.0, 540.0, 810.0, 1215.0, 1822.5, 2733.75] ms, TE = 10 ms) and T2- weighted (TR = 6000 ms, TE = [10.0, 20.0, 40.0, 80.0, 160.0, 320.0] ms) images were simulated by
S(Q)=PD(((1-e^(-TR/T_1 ) ) sin(α))/(1-e^(-TR/T_1 ) cos(α) )) e^(-TE/T_2 ) (1)
with α = 90°. The resolution of images (C_h ) was lowered by setting high spatial frequencies to zero, C_l=Φ(C_h). Maximum likelihood (MLE) estimation [5] was used to obtain the low-resolution qMRI Q_l from these images. High-resolution guide images W were synthesized using S with settings for SPGR T1w (α = 12°, TR = 6 ms, TE = 2 ms) and Spin Echo T2w (α = 20°, TR = 5211 ms, TE = 146 ms). A ResNet-based model [6] was trained to super-resolve low-resolution qMRI by leveraging high-frequency details from guide images, minimizing the sum of:
L_l: Mean squared error between the degraded contrast images and Φ(S((Q_h ) ̂ )) where (Q_h ) ̂ is the predicted qMRI.
L_h: complement of Pearson correlation between the guide images and S((Q_h ) ̂ ) with acquisition settings of W.
The training pipeline is illustrated in Figure 1. Figure 2 presents the results of the trained model on a validation subject.
Table 1 summarizes the average metric gains (HFEN and SSIM) across all validation subjects for each parametric map.
Figure 3 demonstrates the same model applied to low-resolution qMRI acquired with MUPA.
• Figure 3b (bottom) displays T1W and T2 images synthesized from the maps. The visual inspection of a single slice from the model trained on synthetic data demonstrates that PHIRE-Q effectively super-resolves degraded qMRI using high-resolution synthetic weighted images as guides, without requiring high-resolution qMRI during training. This is further supported by evaluation metrics computed for all validation subjects, as reported in Table 1. When applied to low-resolution qMRI from the MUPA protocol, parametric maps improved, approaching the fully sampled version. T1 and T2 maps showed improvements, visually even surpassing the reference though some bias in T1 was introduced. Some artifacts in PD need further investigation. A key indicator of the model's effectiveness is that (Q_h ) ̂ synthesizes images closely resembling the guide images more accurately than those synthesized from Q_l or from the fully sampled Q_h.
This work also explores the novelty of PHIRE-Q: its ability to generalize across different qMRI sequences, which is achieved by using low resolution qMRI as input instead of the original weighted images. During inference this avoids the complexity of the forward physical model of qMRI, including weighted image generation and resolution lowering operator, Φ, which would need non-uniform FFTs for non-Cartesian trajectories as employed in MUPA. We introduced PHIRE-Q, a framework that super-resolves qMRI in a physics-informed, self-supervised fashion without requiring high-resolution ground truth maps. By leveraging the structural detail of high-resolution weighted images and the intrinsic tissue specificity of qMRI, we demonstrated that PHIRE-Q generalizes across a different qMRI sequence and can be used for qMRI super-resolution. Further evaluation on pathological data is required.
Alireza SAMADIFARDHERIS (Rotterdam, The Netherlands), Dirk H.j. POOT, Shishuai WANG, Florian WIESINGER, Stefan KLEIN, Juan A. HERNANDEZ-TAMAMES1,3
13:50 - 13:52
#47892 - PG089 Mapping Tissue-Specific Diffusivity in Gliomas with High b-Value Spherical Tensor Encoding.
PG089 Mapping Tissue-Specific Diffusivity in Gliomas with High b-Value Spherical Tensor Encoding.
Gliomas remain challenging in neuro-oncology, especially mapping internal structure and defining tumor margins. Diffusion MRI (dMRI), with its sensitivity to tissue microstructure, may aid in this, as a reduced apparent diffusion coefficient (ADC) is associated with increased cellularity1. However, ADC is confounded by extracellular free water—both within tumors and surrounding edema, reducing its specificity2. While model-based techniques like free water elimination (FWE)3 attempts to separate tissue and free water post hoc, they rely on assumptions often invalid in tumors. In this work, we investigated whether glioma characterization could be improved by high b-value spherical b-tensor diffusion encoding4, which suppresses free water signal. The method yields maps of the mean diffusivity in tissue (MDT), alongside conventional ADC5. Our aim was to assess MDT maps from gliomas and compare their contrast to ADC maps.
We studied 54 glioma patients (9 astrocytomas, 7 oligodendrogliomas and 38 glioblastomas), classified by histology and IDH mutation status after written and informed consent and permission from the ethical review board. MRI was performed on a 3T-scanner (MAGNETOM Prisma, Siemens Healthineers, Forchheim, Germany), and included structural MRI (T1-MPRAGE with and without contrast, T2-weighted imaging and FLAIR) and diffusion-weighted imaging with 2.3 mm isotropic voxels, spherical b-tensor encoding, and b-values of 0.1, 1.4, 2.0 ms/µm2 in 6, 11 and 16 repetitions, using a research sequence. ADC and MDT maps were calculated as:
D=(log(S(b2))-log(S(b1)))/(b2-b1),
where D is the diffusion coefficient (ADC or MDT), and S(b) is the signal at each b-value. For ADC, b1= 0.1 and b2= 1.4 µm2/ms and for MDT, b1= 1.4 and b2= 2.0 µm2/ms. Regions of interests were segmented using BRATS6 and FreeSurfer7, and covered necrosis, contrast-enhancing tumor, edema and normal-appearing white matter (NAWM). Joint histograms of ADC and MDT across all patients were created for the whole tumor (necrosis and contrast-enhancing regions), edema and NAWM, to analyze covariation patterns between these parameters. Clusters were defined using a Gaussian mixture model. For each patient, the percentage of the tumor voxels belonging to each cluster, the mean MDT and mean ADC were quantified. Patients were grouped by IDH status and compared using Mann-Whitney U-tests. All maps were also inspected for abnormalities beyond the tumor area. Figure 1 shows the distribution of ADC and MDT across all patients in NAWM, edema and tumor. Edema and NAWM displayed single clusters, while tumors displayed three. In cluster 1, MDT was proportional to ADC, suggesting minimal free water contribution. In cluster 2, MDT was lower than ADC, arguably due to free water suppression. In cluster 3, ADC was high but MDT near zero, consistent with voxels dominated by free water. In ~15% of patients, cluster 2 predominated, revealing regions with restricted diffusion not visible in ADC (Figure 1D). Figure 2 shows six patients with tumors dominated by either cluster 1 or 2. The tumor was more conspicuous in the MDT map than the ADC maps. Suppression of free water in cluster 2 revealed regions with low MDT which appeared normal in ADC maps. Figure 3 shows tumor metrics grouped by IDH status. The percentage of voxels in cluster 2 differed significantly (p<0.05) between the groups, while the mean MDT and mean ADC did not. Figure 4 highlights abnormal findings beyond tumors. These consisted of unilateral reductions in MDT, with corresponding hyperintensities in the high-b DWI. One case exhibited a widespread MDT reduction, while the second displayed a narrow reduction extending from the edema. This work investigated a method for mapping the diffusivity in and around gliomas while suppressing free water in the acquisition stage. The additional information provided by MDT were clearest in around 15% of the patients, revealed restricted diffusion not seen in the ADC—likely obscured by free water. In the remaining patients, the MDT provided no additional information about the tumor compared to the ADC, however, the tumor conspicuity was higher in the MDT map than in the ADC map due to free water suppression. The percentage of voxels in cluster 2 was significantly associated with IDH status which in turn relates to tumor type. This may be explained by known association between edema and IDH status8,9. Future work will focus on investigating the potential utility of these findings by investigating the potential utility of MDT beyond the tumor as identified on the morphological maps. High b-value spherical b-tensor encoding enables mapping of tissue-specific diffusivity by suppressing free water at the acquisition stage, avoiding modeling assumptions that for example FWE in tumors. MDT maps reveal restricted diffusion obscured in ADC, enhance tumor visibility, and identify abnormalities beyond tumor margins—offering a robust and assumption-free complement to conventional dMRI.
Cornelia SÄLL (Lund, Sweden), Tim SALOMONSSON, Filip SZCZEPANKIEWICZ, Pia C SUNDGREN, Markus NILSSON
13:52 - 13:54
#46498 - PG090 Optimising diffusion MRI measurements to maximise microstructural feature sensitivity using Monte-Carlo simulations and estimation theory.
PG090 Optimising diffusion MRI measurements to maximise microstructural feature sensitivity using Monte-Carlo simulations and estimation theory.
The choice of diffusion MRI (dMRI) sequence parameters is important for maximising SNR and microstructural sensitivity(1). Whilst past studies on dMRI parameter optimisation have investigated several properties of diffusion encoding schemes(2–5), the choice of sequence parameters remains largely empirical and heuristic(5).
We propose a new optimisation framework combining Monte-Carlo simulations and Fisher Information (FI) to identify a set of dMRI measurements that maximise microstructural sensitivity over a user-defined range of possible values. Using this framework, we demonstrate an optimisation of the pulsed-gradient spin-echo (PGSE) sequence for test substrates of restricted spheres, identifying a combination of measurements that simultaneously minimises the variance of estimates of spherical radius (ranging from 1.5-9.5 µm) and volume fraction (ranging from 0.32-0.52). We additionally perform a preliminary experimental validation using a microbead phantom.
In the context of microstructural imaging, FI is inversely proportional to the minimum achievable uncertainty of an unbiased microstructural parameter estimate (e.g. restriction size) for a given set of dMRI measurements. Mathematically, it computes the microstructural sensitivity using signal derivatives against relevant microstructural features and allows us to combine sequences’ sensitivity to multiple features. To optimise sensitivity, we aim to identify sequence parameters that maximise the signal derivative while maintaining SNR.
Figure 1 illustrates our optimisation scheme. First, we use Monte-Carlo simulations to obtain dMRI signal predictions across a range of discrete values of target microstructural features (θ) and sequence parameters (ϕ). We then estimate derivatives of the simulated signal as a function of θ and interpolate these to form a continuous parameter space. This enables us to calculate the FI matrix and perform an optimisation to identify a measurement set that minimises the uncertainty of the parameter estimates.
We applied our optimisation scheme (Figure 1) to the PGSE sequence for a substrate of randomly distributed restricted spheres (uniform radius), identifying a parameter set that minimised uncertainty on radius and volume fraction estimates. Substrates were simulated using MCMRsimulator(6), simulating 27 substrates with radii ranging from 1.5 to 9.5µm (1 µm interval) and volume fraction ranging from 0.32 to 0.52 (0.1 interval). For each substrate, we simulated 23205 distinct sequence parameter combinations, with gradient amplitude (G) ranging from 0-660mT/m, gradient duration (δ) 0-84ms, diffusion time (Δ) 4-85ms.
We then calculated the signal’s numerical derivative (double-sided) relative to the sphere radius and volume fraction at radii of [2.5, 5.5, 8.5]µm and a volume fraction of 0.42. We subsequently spline interpolated derivatives between sampled points in sequence parameter dimensions using scipy(7). Optimisations were performed for a user-defined number of measurements (2 to 5 for now), where we additionally optimised the relative fraction of time that should be spent sampling each measurement. FI estimates were weighted by T2 decay to account for lower SNR due to longer TE.
For comparison, we also ran optimisations for a single radius and an AxCaliber-style acquisition which sampled sequence parameters using a regular grid. Furthermore, we imaged a set of microbead phantoms with µm-level radii using optimised sequence parameters and AxCaliber acquisition on a Bruker Biospec 7T scanner. We then estimated radius and volume fraction in a single voxel using an interpolated simulated signal dictionary. Figure 2 shows the optimised measurements and AxCaliber comparison with matched total number of repeats. Optimising simultaneously over the radius range converged after 4 measurements. Figure 3 shows that measurements optimised at a single radius have smaller estimation uncertainty near their designated radius, while the measurement set optimised across all radii achieves more consistent estimation uncertainty across the radius range. Figure 4a&b shows that the estimation uncertainty for radius and volume fraction tends to be lower for our optimised sequences compared with the AxCaliber-like acquisition. Table 4c additionally showed our sequence gives estimates that are close to true mean radii overall. We demonstrated that only a few PGSE measurements are theoretically needed to maximise sensitivity to two microstructure features for substrate of restricted spheres, with optimal sequences comparable to previous studies(8). Our proposed optimisation framework can be directly translated to other geometries and dMRI sequences. Going forward, we will further develop analysis pipelines for our acquired phantom data, alongside with a complementary study in ex-vivo rat brains to characterise tissue microstructure. We presented a novel sequence optimisation approach combining Monte-Carlo simulations and estimation theory.
Zhiyu ZHENG (Oxford, United Kingdom), Karla L. MILLER, Mohamed TACHROUNT, Kamila SZULC-LERCH, Sean SMART, Kamila BLACHOWIAK, Benjamin C. TENDLER, Michiel COTTAAR
13:54 - 13:56
#47777 - PG091 Resomapper: a user friendly and versatile pipeline for multiparametric MRI data processing and mapping.
PG091 Resomapper: a user friendly and versatile pipeline for multiparametric MRI data processing and mapping.
Magnetic resonance imaging (MRI) is essential in research and clinical settings, with quantitative MRI (qMRI) improving reproducibility and sensitivity. However, qMRI processing can be challenging, especially for users with limited coding experience. We introduce Resomapper, an open-source, cross-platform tool that integrates well-established processing libraries into a unified, user-friendly workflow, simplifying qMRI analysis and promoting accessibility, reproducibility, and data sharing.
Resomapper is a Python-based tool designed for user-friendly multiparametric MRI processing. It integrates well-established libraries, including Nibabel [1] and SimpleITK [2] for image and data handling, Dipy [3] for diffusion modeling, and others. The software supports T1, T2, and T2* relaxometry, magnetization transfer imaging (MTI), diffusion tensor imaging (DTI), and simple apparent diffusion coefficient (ADC) fitting. To enhance data quality, Resomapper includes preprocessing options such as denoising, Gibbs artifact removal, and bias field correction. Users can process data via an interactive sequential pipeline or an automated JSON-configured workflow, both executable through simple command-line instructions (see Figure 1 for an overview of the workflow). Resomapper ensures compatibility by converting raw MRI data from different formats (DICOM, Bruker, MR Solutions) into the standardized NIfTI format within a BIDS-like structure [4], enhancing reproducibility, scalability, and data management efficiency. As an example of application of the software, we present a brain MRI study carried out on healthy, adult C57BL/6J mice, both males and females (n=34). MRI acquisitions were conducted in a Bruker Biospec 7T system, with a multiparametric MRI protocol including anatomical T2W images, T2 and T2* maps, MTI and DTI. The studies were processed with Resomapper and then co-registered using ANTsPy [3]. Finally, a region of interest (ROI)-based analysis was performed to check for differences between sex and brain areas. The selected ROIs include the cortex (Cx), hippocampus, (HPC), thalamus (Thal), and hypothalamus (HTH). The results of the preclinical MRI studies are shown in Figure 2. Expected differences were found between brain regions in all parameters, and in addition, a small significant sex difference was observed in T2*, being the values found in the thalamus and hypothalamus higher in female mice than in males. This might mean that anesthesia affects differently across sex (T2* is the last acquisition in the MRI protocol, in this case). Moreover, this example serves as a reference for other multiparametric MRI studies in mice, demonstrating the feasibility of using Resomapper for standardized multiparametric qMRI analysis. By integrating multiple processing tools into a single, user-friendly framework, Resomapper simplifies qMRI workflows and facilitates reproducible, high-quality image analysis. Its support for diverse preprocessing techniques, multiple imaging modalities, and standardized data formats makes it a valuable tool for researchers with varying levels of programming expertise. Future work will focus on expanding modality support, improving automation, and enhancing compatibility with clinical datasets.
Raquel GONZÁLEZ-ALDAY, Adriana FERREIRO, Nuria ARIAS-RAMOS, Blanca LIZARBE, Pilar LÓPEZ-LARRUBIA (Madrid, Spain)
13:56 - 13:58
#47910 - PG092 Evaluating the impact of T2 relaxation inclusion in VERDICT model fitting for brain tumors.
PG092 Evaluating the impact of T2 relaxation inclusion in VERDICT model fitting for brain tumors.
The VERDICT model is designed to be fit to diffusion MRI (dMRI) data acquired in tumors using various diffusion times and b-values, enabling the estimation of microstructural parameters such as compartment volume fractions (fX, for compartment X) and cell radius (R) [1]. Recently, an extended model, termed rVERDICT, has been proposed, which incorporates the estimation of intracellular (IC) and extracellular/extravascular (EES/VASC) T2 relaxation [2]. Including T2 relaxation in dMRI models is particularly relevant in clinical settings, where limitations in gradient strengths necessitate longer TEs. Without accounting for these effects, substantial misestimations in parameter values may occur. Although rVERDICT has previously been tested in prostate cancer [2], it has not yet been applied to brain tumors in a clinical setting.
This study aimed to assess the impact of including T2 relaxation in the VERDICT model by comparing results from different TE schemes through in vivo measurements in brain tumors.
Three patients were included in the study, each presenting with a different type of brain tumor: meningioma, oligodendroglioma, and one radiologically diagnosed as glioblastoma. Scans were performed on a 3T Philips MR 7700 system (Best, the Netherlands; release 11.1.0.3) using a 32-channel head coil. Diffusion-weighted imaging (DWI) was conducted using a PGSE protocol across five measurements, with parameters set similar to ref [2]. A dual-echo EPI readout sequence (in-house developed software patch) was used to acquire two echoes, one at the minimum TE (TE1) and one fixed at 140 ms (TE2). Each measurement used specific TE1 (ms), b-value (s/mm²), δ (ms), and Δ (ms) values: respectively, (100,3000,24.8,43.8), (102,1500,25.8,43.4), (61,90,4.7,23.5), (76,500,12.2,31.3), and (82,2000,16.5,33.1), with TR = 5500 ms. A b = 0 image was acquired for each measurement. Anatomical images included T2W FLAIR and contrast-enhanced T1W scans.
The study was approved by the Swedish Ethical Review Authority (ref no 2020-00029).
The rVERDICT model was fit to data within tumor regions of interest (ROIs) with three distinct fitting scenarios: (1) using data for all TEs and including estimation of both T2IC and T2VASC/EES, as detailed in ref [2]; (2) using only the second echo at a fixed TE = 140 ms, with T2 relaxation excluded from the model; and (3) using only the first echo at minimum TE for each measurement, also excluding T2 relaxation. Model fitting was performed as in ref [2] using a deep neural network (DNN) trained on simulated data with added noise consistent with tumor ROIs at b = 0 and TE = 140 ms (SNR = 50). Parameter estimates differed substantially across the three fitting scenarios (Figure 1). Notably, the inclusion of T2 relaxation in the model resulted in higher estimates of fIC and R, while fEES and fVASC were estimated lower. The two scenarios with T2 relaxation excluded showed similar trends to each other. Overall trends were consistent across all tumor types.
Parameter heterogeneity within the tumors varied for the three fitting scenarios (Figure 2). Overall, T2IC values were estimated as much lower than T2VASC/EES, with estimates ranging approximately from 50 – 100 ms for T2IC, compared to 150 – 350 ms for T2VASC/EES (Figure 3). In this study, T2IC was estimated to be substantially lower than T2VASC/EES. Because VERDICT volume fraction parameters reflect signal fractions, a notable bias is therefore expected when T2 relaxation is not accounted for. In protocols with a fixed TE, fIC is expected to be underestimated due to the faster T2 relaxation of the IC compartment. This aligns with the lower fIC observed in this study when T2 relaxation was excluded from the model and a fixed TE was used.
Due to limitations in maximum gradient strength, short diffusion times were only achievable at low b-values, while high b-values required longer Δ. As a result, there was a positive correlation between minimum TE and b-value. This is expected to cause faster signal attenuation with increasing b-values, since TE also increases. This rapid attenuation more closely resembles the behavior of the EES/VASC signal, potentially leading to an underestimation of fIC when T2 relaxation is not accounted for in the model. Again, this is consistent with the lower fIC values observed when T2 relaxation was not included in the model and the minimum TE was used. Incorporating T2 relaxation in the VERDICT model resulted in substantial differences across all parameter estimates and tumor types, yielding estimates that appear more physiologically plausible, with higher fIC and lower fVASC. These findings suggest that including T2 relaxation in the model may improve estimation accuracy, particularly in clinical settings where higher TEs are commonly required. Further validation through studies that include comparisons with histological data is warranted.
Lukas LUNDHOLM (Gothenburg, Sweden), Oscar JALNEFJORD, Mikael MONTELIUS, Mats LAESSER, Thomas OLSSON BONTELL, Alba CORELL, Asgeir STORE JAKOLA, Isabella Maria BJÖRKMAN-BURTSCHER, Maria LJUNGBERG
13:58 - 14:00
#47894 - PG093 Anisotropy of the Myelin Water Signal is influenced by echo spacing and fractional anisotropy.
PG093 Anisotropy of the Myelin Water Signal is influenced by echo spacing and fractional anisotropy.
Myelin Water Imaging (MWI) correlates with myelination of white matter (WM) by exploiting the distinct T2 relaxations times of myelin water (MW, approximately 10 ms at 3 T) compared to intra/extracellular water (IECW, 70-90 ms at 3 T) [1]. Although MWI is well-established, recent studies have demonstrated significant orientation-dependent effects in WM depending on the angle between WM fibers and the main magnetic field B0 [2]. This anisotropy is primarily attributed to magnetic susceptibility differences in myelinated axons and dipole-dipole interactions [3]. Importantly, the choice of MWI pulse sequence and repetition time (TR) influences the magnitude of these orientation effects [2]. MWI anisotropy has also been observed in unmyelinated neonatal WM, suggesting that additional microstructural factors may contribute to this phenomenon [3]. A key technical challenge in MWI is that the short T2 relaxation time of MW closely matches the first echo time (TE) and echo spacing (ΔTE) of many standard MRI sequences, which may confound the assessment of orientation-dependent effects. This study investigates the effect of ΔTE and fractional anisotropy (FA) on the orientation dependency of MWI.
Five healthy subjects were scanned at 3 T using a multi-echo CMPG sequence with multiple echo spacings ΔTE = 8, 9.6, 12, 16 ms (TR = 1100 ms) for MWI. Fiber angle and FA were estimated using diffusion tensor imaging (DTI; b = 700 s/mm^2, 60 directions, ΔTE = 60.42 ms, TR = 4127.851 ms). T1 weighted images for tissue segmentation were acquired using a MPRAGE sequence (ΔTE = 4.52 ms, TR = 9.879 ms, flipangle = 8.06°).
MWI was computed using DECAES [4], which fits the signal to the T2-decay curve multi-exponentially using NNLS resulting in a voxel wise T_2-Distribution. From these several maps are extracted: the global geometric mean (ggm), the IECW and MW R2 along the IECW fraction and MWF. The T2 cutoff of 25 ms was used to separate short from medium T2 components. The fiber angle was calculated as the angle between the first DTI eigenvector and the direction of the main magnetic field. Figure 1A shows T2 spectra for varying ΔTE. As ΔTE increases, the MW peak shifts toward longer T2 times, whereas the IECW peak shifts in the opposite direction. Notably, increasing ΔTE results in a marked decrease of the MWF, from 5.2% at ΔTE = 8 ms to 2.7% at ΔTE = 16 ms (Figure 1B), partly due to the fixed T2 cut-off at 25 ms, which remains a robust choice as the MW peak consistently falls below it.
WM voxel counts (FA ∈ [0, 1]) generally increase with fiber angle (Figure 2), low (FA ∈ [0, 0.5]) and high FA (FA ∈ [0, 0.5]) reflect that, tough high FA include fewer voxels.
Figure 3 shows the orientation dependence of the ggm R2, IECW R2, and MW R2 for each FA range. All three parameters exhibit orientation dependence, with IECW R2 showing the weakest and MW R2 the strongest effects. IECW and ggm demonstrate minimal sensitivity to changes in ΔTE as shown by the minimal offset between the curves. In contrast, MW R2 decreases strongly with increasing ΔTE as well as its orientation dependency. Of note, the minimum of the ggm curve aligns closely with the magic angle, while the minima for the IECW and MW R2 occur at lower fiber angles, as detailed in Figure 1 and Table 1.
Both, IECW and ggm exhibit similarly low anisotropy indices (Table 1), with IECW’s curve shape remaining consistent across ΔTE and FA. MW shows a significantly higher anisotropy index, up to ten times that of IECW at high FA. Our findings demonstrate that both the MW signal and its anisotropy are highly sensitive to the ΔTE of the MRI sequence. As ΔTE increases, the accuracy of MW signal estimation declines, resulting in a systematic underestimation of the MWF. The influence of ΔTE is particularly evident in the MW orientation curve, where a pronounced peak emerges around 20 degrees at the shortest ΔTE. This peak diminishes and nearly disappears as ΔTE increases beyond the optimal range for capturing the MW signal. Although the MW signal exhibits substantial orientation dependence, it does not strictly follow the classic magic angle pattern. In contrast, the ggm, which reflects an average of MW and IECW contributions, displays a clear signal minimum at the magic angle. In contrast, the IECW curve remains stable, showing no significant dependence on ΔTE or FA range. In summary, our results underscore the necessity of carefully optimizing ΔTE for more accurate quantification of MWI and reliable assessment of its orientation dependence, particularly in regions with high FA. The strong orientation dependency and rapid decay of the MW signal make it especially susceptible to variations in ΔTE and FA, whereas the IECW signal remains stable across different acquisition settings. These insights provide a foundation for refining MWI protocols and highlight the potential for FA-based corrections to minimize orientation-related biases in myelin water imaging.
Juliana FELBER (Innsbruck, Austria), Manuel BAUER, Alexander RAUSCHER, Christoph BIRKL
14:00 - 14:02
#47711 - PG094 Multi-Contrast Super-Resolution Reconstruction Using Implicit Neural Representations for Robust Brain T₂ Mapping based on 2D SS-FSE series in the Presence of Severe Motion and Incomplete Data.
PG094 Multi-Contrast Super-Resolution Reconstruction Using Implicit Neural Representations for Robust Brain T₂ Mapping based on 2D SS-FSE series in the Presence of Severe Motion and Incomplete Data.
Quantitative T2 relaxometry mapping offers valuable biomarkers for early brain development [1]. Pioneering work in fetal MRI at 1.5T uses thick 2D T2-weighted (T2-w) SS-FSE slices at high field across multiple echo times (TEs) and orientations for super-resolution reconstruction (SRR) and mapping of the T2 decay through an exponential fit [2]. A major limitation is the need for at least three orthogonal stacks per TE before SRR [5], often corrupted by unpredictable fetal motion, especially under limited scan time in clinical settings.
Feasibility of in vivo T2 mapping at lower fields (0.55T, 64mT) has also been explored in adults and phantoms [3,4] using interpolation instead of SRR. Extending this to the fetal brain could democratize early development research in low-resource settings.
We propose a joint multi-TE SRR using implicit neural representation (INRs) [6]. Our framework, based on [7], models anatomy as a continuous function [6–9] and allows information sharing across contrasts [10, 11]. This joint TE SRR approach could enhance motion robustness and reduce acquisition time by exploiting data redundancy across TEs, without affecting quantitative metrics like T2 decay.
We use simulated and in vivo datasets.
-5 T2-w fetal brain stacks at 25, 27, 30, 33, and 35 weeks GA were simulated using FaBiAN [12] with motion and signal drop-out at 1.5T. Image resolution is 0.8 × 0.8 × 4.5 mm³. Ground-truth (GT) T2 values are 280 ms in white matter (WM) and 200 ms in gray matter (GM) [5]. Images are generated at TEs of 220, 500 and 690 ms, TR of 12ms. Additionally, a high-resolution (0.8 mm isotropic), motion-free volume is generated as a GT for SRR evaluation, from which GT T2 values are estimated to assess T2 without SS-FSE-related bias [2].
-5 T2-w in vivo adult brains scanned at 0.55T (Siemens Free.Max) with TEs of 114, 220, and 290 ms with a total acquisition time of 16.5 minutes [3]. Image resolution is 1.1 × 1.1 × 4.5 mm³. Synthetic fetal-like motion and signal drop-out are applied [12, 13]. GT SRR and T2 values are derived from initial volumes via resampling to 1.1 × 1.1 × 1.1 mm³, denoising and trilinear interpolation [3]. WM and GM tissues are segmented using SynthSeg [15].
3D SRR: inspired by [7,9], we perform motion correction and slice-to-volume reconstruction (SVR) in a self-supervised manner, with no reliance on pretrained models unlike prior algorithms [9,16]. The resulting output resolution is 0.8 × 0.8 × 0.8 mm³ for the FaBian data and 1 × 1 × 1 mm³ for the Adult data.
We compared 2 variants (Fig. 1):
-Single-contrast INR (Single-C): trained independently per TE.
-Multi-contrast INR (Multi-C): jointly trained across TEs, using a single network with all the stacks as input and each TE as a separate output dimension.
Evaluation: Each variant is evaluated for SRR and T2 fitting quality under the following conditions:
-Full acquisition: 3 stacks per TE (Experiment A).
-Reduced acquisition: 2 stacks per TE (Experiment B).
For both variants, network hyperparameters were kept constant.
T2-mapping: We first applied brain extraction [17], rigid co-registration across TEs [18] and voxel-wise T2 exponential fitting assuming Gaussian-distributed noise [4]. Average T2 values (in ms) were computed in WM and in deep GM (DGM), and statistical significance of the 2 variants was assessed using a two-sample t-test. Multi-C SRR demonstrates superior quality compared to Single-C SRR in both adult experiments (A and B), as reflected by higher SSIM [19] values (Table 1). In FaBian, the SSIM scores between Multi-C and Single-C are similar, although more image artefacts are visible in Single-C (Fig. 3).
Mean T2 relaxometry values in WM and DGM are accurately estimated when all 3 orthogonal orientations per TE are used (A) (Table 1). When only two orientations per TE are used (B), Multi-C maintains a relatively low mean error in WM compared to Single-C and similar in DGM (Fig. 2) but the standard deviations of T2 values remain high across both variants (Table 1). Statistical testing confirms a significant difference between Reduced SC and MC in both adults (p=0.05) and FaBian (p=0.04) WM. Multi-contrast SRR of the same brain propagates the same geometrical features to other contrasts, filling missing structural information at a given TE, reducing visual artefacts in SRR volumes and T2 maps (Fig. 1 and Fig. 3). This improves structural consistency while preserving quantitative T2 values. While mean T2 values in WM and DGM remain robust, even when one stack per TE is missing, the high standard deviation in reduced-data settings indicates suboptimal inter-TE information exchange. Future work will integrate physical priors to enhance cross-contrast consistency, tests on larger datasets, including validation on in vivo fetal brain scans. By integrating multiple TEs in a multi-contrast SRR framework, we achieve robust T2 mapping from fewer SS-FSE acquisitions than single contrast methods, towards clinically feasible fetal brain relaxometry.
Busra BULUT (Lausanne, Switzerland), Maik DANNECKER, Steven JIA, Margaux ROULET, Vladyslav ZALEVSKYI, Thomas SANCHEZ, Jean-Baptiste LEDOUX, Guillaume AUZIAS, François ROUSSEAU, Daniel RUECKERT, Meritxell BACH CUADRA
14:02 - 14:04
#46172 - PG095 Optimized ZTE Quantitative Magnetization Transfer Imaging.
PG095 Optimized ZTE Quantitative Magnetization Transfer Imaging.
Quantitative Magnetisation Transfer (qMT) MRI has been researched for decades, especially for indirectly imaging myelin. Recently, a protocol has been described which can extract maps for 9 model parameters in clinically acceptable scan times [1].
While the method provided in [1] provides us with a "gold standard", we chose to investigate the acquisition of qMT MRI using a ZTE readout. ZTE uses hard, non-selective readout pulses with flip angles [0 - 4°], which limits the SNR and the possible optimisation of the sequence [2], and may therefore limit precision and efficiency. Moreover, information about the transverse relaxation is difficult to obtain.
ZTE is, however, a desirable readout for its near-silent operation and its sensitivity to short T2 species which potentially allows direct access to the myelin signal. Due to hardware limitations, even with ZTE, direct myelin imaging has been limited to systems with very high-performance gradients so we propose instead to use ZTE-based qMT imaging, focusing on its reduced noise levels.
We investigate extensions to the previous ZTE qMT imaging work [3] to which may provide more precise of mapping of a greater number of model parameters.
We used a local implementation of the ZTE sequence on a 3T Premier system (GE Healthcare). Our readout achieves incoherence at each time point along the readout by sampling a hemispherical portion of k-space in each segment and rotating the trajectory by 2D golden means after having sampled the opposite hemisphere in the following segment [4]. This also allows us to minimise unwanted refocusing effects.
Such incoherent sampling is particularly important for the subspace-based reconstruction used in this work to extract the magnetisation dynamics at each spoke along the readout. The subspace reconstruction was performed using Riesling [5].
After testing several combinations of preparation flip angles and spokes-per-segment (SPS), the final sequence was determined heuristically in order to optimise encoding of the MT-model parameters. In line with previous literature [1], we chose to use a short (1ms) inversion pulse before the first ZTE segment. The following two segments had preparation pulses of duration 1ms and flip angle (FA) 270°, while the subsequent two had preparations of duration/FA = 1ms/15°. This train of prepared ZTE readouts was repeated until the Nyquist sampling criterion was satisfied. The duration of each of these ZTE trains is about 4.2 seconds for SPS = 384, which ensures some recovery of the longitudinal magnetisation during the train. In line with previous work on 'pulsed' ZTE qMT [3], we also varied the readout flip angles to increase T1-weighting: the first segment in the train used a 4° flip angle, the second 4°, then 2°, 4° and 2°.
The whole sequence is conveniently modeled using the linear approximation to the Generalised Bloch Model [7]. The ZTE steady state can be solved for in homogenised form, as in [6]. A Python simulation was used to calculate the subspace basis, as well as to derive model parameter maps by least squares fitting.
Other sequence parameters: matrix size = 200, FOV=240cm and TR=2.2ms (BW= ±25kHz). To increase SNR, we collected 5 times the number of samples required to satisfy the Nyquist criterion. This resulted in an overall scan time of about 8 minutes.
While we reconstructed the data with a subspace spanning typical model parameters [1], we constrained our fits to only four of these. Parameter maps were obtained for the macromolecular fraction f_s, the T1 of the free pool T1f, PD and B1 when fixing the exchange rate Rx to 14 s^-1 and T1s=0.35s^-1 [1], Figure 1.
The results show good image quality for all maps except for T1f. The latter often hits an upper bound and is heavily influenced by noise.
Constraining the T1f = T1s = T1app leads to visually more appealing results in which the T1 estimates get biased towards Rx and f_s [1], Figure 2. The method proposed here provides a good-quality map of the macromolecular fraction, which we intend to assess for its clinical utility. We note from a visual inspection that the values in the map are higher than the current literature [1], possibly due to biases arising from the fitting constraints which will be investigated.
Further research should investigate ways to disentangle T1f for effective estimation. To this end, we aim to further optimise the sequence, using the Cramer-Rao bound (CRB) as a metric. The CRB can be conveniently extracted by modifying our current simulation framework.
Further work will also assess the trade-off between SNR, number of samples collected and reconstructed voxel size to improve estimation while still retaining clinically acceptable scan times and image quality. This work presents a method to obtain estimates of f_s, (T1app), B1 and PD in a single ZTE scan. This method could be expanded to obtain additional, precise, MT parameter estimates.
Oliver PINNA (London, United Kingdom), Tobias WOOD, Gareth BARKER
14:04 - 14:06
#47746 - PG096 Mapping cerebrovascular reactivity using ASL and BOLD-fMRI and its relationship with baseline perfusion.
PG096 Mapping cerebrovascular reactivity using ASL and BOLD-fMRI and its relationship with baseline perfusion.
Cerebrovascular reactivity (CVR) measures perfusion changes in response to vasoactive stimuli, reflecting brain vascular health [1]. BOLD-fMRI is used to map CVR by providing a surrogate of cerebral blood volume (CBV) changes. However, recent studies show that CVR-BOLD strongly depends on baseline CBV, particularly in white matter (WM) and densely vascularized regions [2-5]. In contrast, Arterial Spin Labeling (ASL) can assess CVR in terms of cerebral blood flow (CBF) (CVR-ASL) changes, and is therefore not affected by baseline CBV [2,3], especially in vascular areas [2-6]. Most studies investigating the relationship between CVR and baseline CBF (bCBF) has been previously related to CVR-BOLD [6-9]. So far, only one study has examined the relationship between bCBF and CVR-ASL using pulsed ASL (PASL), instead of the recommended 3D GRASE pseudo-continuous ASL (pCASL) sequence [6]. Here, we aim to characterize CVR-ASL and CVR-BOLD and their relationships with bCBF in gray matter (GM) using a simultaneous pCASL and BOLD acquisition [10].
Data Acquisition: 17 healthy subjects (19-37yrs, 13F) were scanned on 3T Siemens PrismaFit MRI with a 64-channel head RF coil and a dual-acquisition pCASL-BOLD sequence [10] (TR=7s and voxel size=3.5x3.5x5.0mm3). pCASL data were acquired at TE=31ms, with a 3D GRASE readout and background suppression (LD=1.8s, PLD=1.8s, 63 repeats). BOLD-fMRI data were acquired using a 2D multi-echo (ME) EPI at three TEs of 17, 45, and 73ms (here, only data of TE=45ms was used). Both metrics were collected during a hypercapnia (HC) gas challenge that included 5min of baseline, 5min of inhaled gas mixed with 5% CO2, and 5min of baseline.
Data Analysis: Volume realignment was done with FSL. For BOLD data, spatial smoothing (FWHM=5.25mm) and high-pass temporal filtering (cutoff=100s) were applied.
bCBF maps (ml/100g/min) were obtained from the first 5min baseline and the HC using BASIL, by averaging paired control-label subtraction images with spatial regularization and voxelwise calibration using the M0 tissue image. CVR-ASL (%/mmHg) was computed as in [5].
For BOLD-fMRI, a GLM was performed considering the PetCO2 signal [11], a label-control waveform, motion parameters and outliers as regressors, and CVR-BOLD (%/mmHg) was obtained as in [11].
Maps were registered to MNI space and thresholded: bCBF from 5th percentile to 100ml/100g/min for valid physiological ranges; CVR-ASL from 5th-97th percentile to remove potential outliers, and CVR-BOLD from 0 to max, or the 95th or 90th percentile, to minimize macrovascular confounds.
FSL's Randomise was used to perform voxelwise group-level permutation testing between CVR-ASL and unthresholded CVR-BOLD maps.
For pairwise relationships in GM (segmented with FSL’s FIRST), the bCBF or CVR-ASL maps (independent variable) were stratified into 20 bins, and the mean value of the dependent variable (CVR-ASL or CVR-BOLD) was computed across the corresponding voxels [8]. R’s Stepwise linear mixed model analysis was performed considering linear and quadratic terms (fixed factors), and subject as a random effect (CVR-BOLD ~ bCBF + bCBF^2 + (1|Sub)) [12]. Fig.1 shows the bCBF, CBF-HC, CVR-ASL and CVR-BOLD maps. CVR-ASL presents lower GM/WM contrast and reduced values in arterial regions compared to CVR-BOLD.
Fig.2 compares CVR-ASL and CVR-BOLD, highlighting significant differences mainly in WM and in highly-vascularized regions. These results are consistent with a strong CBV influence on CVR-BOLD, with lower values in WM due to low CBV (vs to GM) and higher values in vascular regions due to elevated CBV.
Fig.3 plots the pairwise relationships between bCBF, CVR-ASL and CVR-BOLD, with CVR-BOLD thresholded at various levels. The results show a quadratic relationship between bCBF and CVR-ASL. By spatially mapping the two components of the correlation curve, we found that the initial positive correlation corresponded to regions likely affected by motion, while most of the GM showed the expected negative correlation (Fig.4). The association between bCBF and CVR-BOLD tends to be more linear and positive as the threshold increases. This is expected as bCBF is unaffected by CBV and by eliminating this confound, both measurements become more associated. There is no relationship between CVR-ASL and CVR-BOLD. Our findings support the literature [6-9] showing that CVR measured with BOLD-fMRI is confounded by CBV, as such CVR-ASL and CVR-BOLD represent different measures. The correlation between bCBF and CVR-BOLD, thr90 is consistent with our previous findings [7], albeit with more variability likely due to the larger partial volume effects and larger voxel sizes or differences in acquisition, such as the use of a segmented pCASL image, which offers higher quality in ASL images. Additionally, we find a negative correlation between bCBF and CVR-ASL, as expected since CVR-ASL is normalized by bCBF. CVR-BOLD is strongly influenced by CBV, whereas CVR-ASL can be used as a more direct measure of CVR.
Catarina DOMINGOS (Portugal, Portugal), Inés CHAVARRÍA, César CABALLERO-GAUDES, Patrícia FIGUEIREDO
|
Salle 120 |
|
"Thursday 09 October"
Added to your list of favorites
Deleted from your list of favorites
G11
13:30 - 15:00
Poster 1
FT1 - Poster qMRI technology | FT3 - Poster Quantitative MRI | FT2 - Poster Methods | FT2 - Poster Toolboxes and datasets
13:30 - 15:00
#47666 - PG223 Using Magnetic Resonance Elastography (MRE) to characterize the biomechanics of the microenvironment of a HepG2 spheroid.
PG223 Using Magnetic Resonance Elastography (MRE) to characterize the biomechanics of the microenvironment of a HepG2 spheroid.
The extracellular matrix (ECM) regulates biological pathways crucial for homeostasis [1]. n solid tumours, ECM–cancer cell interactions alter the microenvironment, influencing tumour progression, metastasis, and treatment response [2] [3] [4] [5]. Significant the Tumoural-Micro-Environment (TME) alterations include excessive accumulation of type I collagen and increased crosslinking, leading to higher stiffness [1] [3].
Magnetic Resonance Elastography (MRE) is widely used to assess tissue biomechanics—commonly in liver fibrosis staging [6]— and is gaining interest for predicting chemotherapy response [7].
Biomechanical modifications of the ECM in the context of cancer are currently receiving significant attention both in vitro and in vivo [5] [8] [9]. So far, MRE on tumour organoids/spheroids has never been investigated [9]. In the context of tailored medicine, developing a 3D culture model, from patient’s biopsy samples, that is predictive to their response to treatment could be relevant. Here we use Magnetic Resonance Elastography (MRE) to locally characterize the biomechanics of a cancer cell spheroid within a collagen matrix.
Matrix preparation
Rat tail collagen type I was mix with ribose (200mM final concentration) and allowed crosslinking for 30 minutes at 4°C. Glycated collagen solution was mixed with OptiMEM, HEPES, fibronectin and neutralized with 1M NaOH (collagen final concentration: 3 mg/mL).
Spheroids generation and embedding
HepG2 cells were grown in 2D until 60% confluency. After trypsinization, 5000 cells/well were seeded in a U-bottom 96-well plate and cultured for 2 days. Then, spheroids were embedded in the collagen matrix: round shaped spheroids were selected with a microscope and mixed to the collagen matrix before plating. The collagen gelled at 37°C for 30 minutes before adding culture media. Additionally, 3% agar gels were prepared from which agar phantom organoids samples were cut into 500 microns pieces. These agar organoids were equally embedded in the collagen matrix. 200uL droplets of hydrogels were plated in a 35mm petri dish, resulting in a dome geometry of 10mm x 1mm.
MRE acquisition and post processing
Data were acquired on a 7T MRI preclinical scanner, using a spin echo MRE sequence with 35 microns resolution in the readout direction (upward direction represented by the red arrow in Figure1), 0.3mm in phase-encoding direction, and 0.5mm slice thickness. Shear waves were induced contactlessly from the bottom of the petri dish, generating planar waves in one spatial direction (Figure1B&Figure2B). HepG2 and agar spheroids were localized within the magnitude image (Figure 1C, D, Figure 2 C, D). Figure 3 shows how the biomechanical properties of the spheroids with their microenvironment were quantified: from the representation of the sine function (Figure 3B) of the wave propagation along the red line of the Figure 3A, sine/cosine function were locally fitted to their imaginary and real part of the wave field using an exponential suppression. The fitting was segmented in three parts: 0-0.6-1.2-1.8mm (Figure 3). Thereby, stiffness could be quantified within a perimeter of 0.9mm laterally to the spheroid and until ~1.8mm longitudinally covering the entire matrix (Figure1D&Figure2D). Figure 4A shows a wave velocity within the tumour spheroid of 0.34 m/s, slowly decreasing to 0.24 m/s within a vicinity of 600 microns (scale C1 showing the wave velocity in m/s) until reaching its nominal value, with increasing the distance to the HepG2 spheroid (scale C2, showing the relative shear wave velocity normalized to HepG2 spheroid). This microenvironment modulation is not seen in the agar spheroid (Figure 4B), where the shear speed drops immediately to its nominal value. Shear speed maps (Figure 4) indicate a high modulation of the tumoural spheroid microenvironment after 2 weeks of culture within a radius of 600 microns. This modulation is shaped as a stiffness gradient around the HepG2 spheroid. In contrast, shear speed dramatically drops around the agar spheroid. Stiffening of the environment is a well-established hallmark in cancer progression, primarily driven by the crosstalk between tumour cells and the TME, which involves extensive remodelling of the ECM and overexpression of collagen [3] [4] [5] [10] [11]. These results go hand in hand with recent results where tissue mechanics is capable to identify very early during neoadjuvant chemotherapy in breast cancer resistance due to changes in biomechanics [7]. Our work has investigated the feasibility of MRE on in vitro models. Here we present a concrete application of micro-MRE on a 2 weeks tumoural spheroid. These encouraging results and the lack of non-invasive methods [9] [12] [13] to investigate biomechanics in vitro motivate us to improve micro-MRE and apply it to the field of tailored medicine with transposable biomarkers.
Marguerite DUCAMP (London, United Kingdom), Gabrielle MANGIN, Asma BOUMAZA, Anne-Sophie VAN SCHELT, Maddy PARSONS, Ralph SINKUS
13:30 - 15:00
#47798 - PG224 Optimization of frequency-modulated QTI-sequences with spiral readout to investigate restricted diffusion for 7T MRI.
PG224 Optimization of frequency-modulated QTI-sequences with spiral readout to investigate restricted diffusion for 7T MRI.
Q-space trajectory imaging (QTI) [1] estimates the diffusion tensor distribution from multiple compartments [2] using spherical (STE), linear (LTE) and planar (PTE) b-tensor shapes to assess anisotropy [3-4]. The encoding spectrum should contain similar frequencies across axes to ensure restricted diffusion measurement at the same diffusion time for each axis and across encodings as their combination should result in a diffusion tensor distribution, specific for one diffusion time [5-7]. To achieve the desired b-tensor shape and encoding spectrum while simultaneously accounting for gradient system limits prolongs the diffusion encoding gradients. This is especially challenging for measurements of white matter tissue at ultra-high field due to the faster T2 signal decay.
In this work, the diffusion gradients of three established QTI methods were optimized for microscopic anisotropy measurements at 7T accounting for shape and encoding spectrum of the b-tensor. These methods were combined with a single shot spiral readout to minimize TE. In vivo QTI-metric maps of the three methods were compared with regard to encoding spectra differences, which were expected to result in metric maps at different diffusion times.
One healthy subject was measured on a MAGNETOM 7T Plus scanner (Siemens Healthineers AG, Germany) with a 32/1 ch receive/transmit coil (Nova Medical), and a maximum gradient strength of 70mT/m.
Three diffusion waveforms were inserted into a single shot spiral spin-echo sequence [8] implemented with Pulseq [9] (Figure 1A). Diffusion waveforms were designed according to the qMAS [10-11], gDOR [6-7] and FAMEDcos [11-12] approaches, which probe different encoding spectra [6-7], and were optimized in the following ways: qMASmod [11] (Figure 2A) was modified with additional correction factors considering a finite gradient slew rate ensuring STE. Using MATLAB’s “fmincon” [13-14], STE waveform parameters were optimized (Figure 1B) to minimize TE with regard to scanner limits and two (one) STE gradient axes were used for PTE (LTE). gDORrefoc (Figure 2B) was adapted from Ref. [6-7]. The one-dimensional gradient g1D(t) [6-7] was optimized using MATLAB’s “generic algorithm” [13,15] leading to a spin-echo with minimal TE (Figure 1C). The double oriented frequency ratio n and the total angle of rotation ΔΨ2 [6-7] were optimized to minimize frequencies near 0Hz. PTE and LTE were optimized separately and STE was excluded to reduce TE. For FAMEDcos [11] (Figure 2C) sinusoidal gradients were exchanged with trapezoidal gradients for STE [12] reducing TE and two (one) STE gradient axes were used for PTE (LTE).
Image reconstruction [16] was done with the PowerGrid toolbox [17] using an expanded encoding model [18], combining B0 and coil sensitivity maps with field camera data (Skope, Zürich). QTI-metric maps were computed on preprocessed data with the QTI± algorithm [19-21]. The metric maps were compared to each other within the splenium (parallel fibers) and the Anterior Corona radiata (ACR) (crossing fibers) to investigate encoding spectra differences of the three methods. Therefore, metrics as well as FSL atlases [22-24] of the regions, were registered to an MP-RAGE image. Violin plot statistics, voxel-wise differences and Cohen’s d [25] were computed. qMASmod resulted in higher µFA and lower Cmd across the white matter compared to gDORrefoc and FAMEDcos (Figure 3).
The comparison between qMASmod and the other approaches in Figure 4 showed a high effect of Cohen’s d (d>0.8) with large voxel differences for µFA and Cmd in the ACR and a slightly reduced effect in the splenium. In contrast, gDORrefoc and FAMEDcos led to lower d in these cases. The MD and FA in the splenium led to d<0.2 (low effect) for all cases with differences centered around zero, whereas qMASmod led to lower MD values and higher FA values in the ACR (d≈0.5). The in vivo measurements showed that QTI-metric maps can be obtained at 7T by combining optimized diffusion gradients with a spiral readout minimizing TE. A maximum b-value of 1400 s/mm² was chosen instead of 2000 s/mm² [3] to not increase TE even further, but still resulting in reasonable anisotropies [1]. The difference of µFA and Cmd in the splenium and ACR could be explained by the difference of the encoding spectra. While qMASmod measures QTI-metrics at very long diffusion times leading to higher µFA and lower Cmd, FAMEDcos and gDORrefoc measure the metrics at shorter diffusion times. These results agree with the literature [6-7]. The difference is more visible for the region with many crossing fibers (ACR) compared to the region with many parallel fibers (splenium) and not that strong for the macroscopic metrics (MD, FA), at least in the investigated regions. QTI-waveforms were optimized and inserted into a spiral spin-echo sequence to obtain feasible acquisition times at 7T. The variation in QTI-metrics across sequences suggests that encoding spectra affect metrics due to different diffusion time.
Svenja NIESEN (Bonn, Germany), Marten VELDMANN, Tony STÖCKER
13:30 - 15:00
#47558 - PG225 Clinical cardiac T1rho mapping.
PG225 Clinical cardiac T1rho mapping.
Myocardial T1 and T2 mapping sequences have been widely used in the past decade to better assess tissue scar and oedema. A novel technique called T1rho mapping had been developed recently (1) to study low frequency phenomenon such proton exchange between water and macromolecules. It is suggested that T1rho would be able to provide contrast between myocardium and interstitial fibrosis and other heart diseases involving large molecules. The key benefit would be in the end to provide myocardial lesion detection and quantificatione without the need of contrast agent injection.
Two patients underwent standard MR clinical procedure on a 1.5T Sola MRI (Siemens Healthineers, Erlangen) consisting of conventional cines, T1, T2 mapping, dynamic perfusion and delayed enhancement protocols. A stack of T1rho slices acquired under breath-hold in short axis view was added to the protocol before contrast agent injection. The T1rho sequence parameters were: FOV 360x290, voxel size 2.5x1.9x8mm, T1rho preparation module used 4 Spin-Lock pulses and 2 refocusing pulses. Myocardial T1rho maps were generated using elastic coregistration and a 2-parameter curve fitting process using five different Spin-Lock times: 0, 10, 20, 35, 50ms. On the first patient with suspected acute myocarditis, as shown on fig.1 there is a very good agreement between delayed enhancement images and T1rho maps, with T1rho values elevation in the area of LGE (78 msec vs. 45 msec). On the second patient with suspected cardiomyopathy (Fig. 2), there is a more discrete enhancement on both delayed enhancement and T1 rho map on the inferolateral wall. T1rho values increase from 44 msec in normal tissue vs. 55msec in abnormal myocardium. T1rho maps exhibit good correlation with delayed enhancement images. This new contrast seems to be very sensitive to either water content or tissue scar. As questions arise whether gadolinium-based contrast agent can be harmful to human health and environment, this technique provides a useful tool for a number of patients (children, pregnant women, patients with renal failure). The relative low resolution compared to a standard delayed enhancement protocol in the other hand could be a drawback for systematic use.
Aurelien MONNET (Nantes), Karine WARIN-FRESSE, Kalvin NARCEAU, Aurelien BUSTIN, Thomas TROALEN
13:30 - 15:00
#47925 - PG226 Modeling Early Cortical Development in Human Organoids: Time-Dependent Mean Diffusivity as a Signature of Cell Body–Extracellular Water Exchange.
PG226 Modeling Early Cortical Development in Human Organoids: Time-Dependent Mean Diffusivity as a Signature of Cell Body–Extracellular Water Exchange.
Cortical development shapes cognitive function and brain health, with abnormalities linked to neurological disorders [1,2]. In early childhood (1.5–6 years), associative neuron maturation drives key cognitive shifts [3]. Studying the late fetal or preterm period, marked by dendritic differentiation and neuronal aggregation, remains challenging in vivo. Brain organoids derived from induced pluripotent stem cells (iPSCs) offer a promising in vitro model, replicating key developmental features while bypassing ethical concerns [4]. Here, we used diffusion MRI (dMRI) to assess microstructural maturation in cortical organoids generated from healthy donor hiPSCs (NeuroNA Human Cellular Neuroscience Platform). We applied diffusion tensor and kurtosis imaging metrics (DTI/DKI) [5] to evaluate maturation [6] and examined diffusion time sensitivity, highlighting organoid-based dMRI as a non-invasive method for studying early cortical development.
Nine brain organoids (~2 mm) were cultured for 4.5 months and combined into three assembloids (~4 mm). Organoids were fixed in 4% paraformaldehyde for up to 2 hours (based on size and ~1 mm/h diffusion rate), with swirling for uniform fixation, then washed in PBS and stored. Assembloids were scanned at CIBM (Lausanne, Switzerland) using a 9.4T Bruker MRI CryoProbe, stabilized in agar within syringe holders. Diffusion MRI was acquired with a PGSE EPI sequence using b-values [1, 2, 3.5, 5, 7] ms/µm², directions [12, 16, 24, 30, 40], and Δ = [15, 26, 38] ms with δ = 4.5 ms; four b=0 images per Δ. Other parameters: TE/TR = 54 ms/2.4 s, 0.2-mm resolution, 0.4-mm slice spacing; 50 min scan per Δ. Data were denoised with Patch2Self, corrected for Gibbs ringing (DIPY), and adjusted for susceptibility, eddy currents, and motion (FSL topup and eddy)[8]. DTI (b=1 ms/µm²) and DKI fits were done per Δ using DIPY. MD time-dependence was analyzed with the CEXI model [9], simulating exchange between intra- (spheres) and extracellular compartments using synthetic b=1 ms/µm² data. Nine cerebral organoids were successfully cultured to 4.5 months. Figure 1A shows a set of three organoids assembled for imaging. The dMRI protocol yielded high-quality signals across all samples, with b=0 and b=7 ms/µm² images shown in Figures 1B and 1C. Based on visual contrast, inner and outer regions were manually segmented. The DKI model was applied separately to datasets acquired at different diffusion times (Δ). Figure 2 shows representative FA, MD, and MK maps for Δ = 15 ms and Δ = 38 ms. FA was generally low and decreased with increasing Δ, with slightly higher values in the outer ring. MD was higher in the inner region and increased with Δ in both regions. MK was elevated in the inner ring and decreased with diffusion time. Voxelwise distributions (Figure 3A) confirmed these regional trends. Figure 3B illustrates a clear increase in mean MD with Δ, consistent across DTI and DKI estimates. To explore the unexpected rise in MD—opposite to the typical decrease seen in white matter [10]—we performed simulations using the CEXI model. With impermeable membranes, MD remained constant or decreased with Δ (Figure 4A). Introducing membrane permeability led to an increase in MD, with the extent depending on cell radius and permeability (Figures 4B and 4C). Our simulations suggest that the increased MD in real data is a hallmark of transmembrane water exchange. Across all samples, FA remained low (<0.15), indicating poor microstructural organization. The outer ring showed slightly higher FA than the inner zone, suggesting early development of elongated pyramidal and callosal neurons, as well as astroglial processes among rounded progenitors. At longer diffusion times, FA declined further due to increased permeability, allowing water to move across aligned and non-aligned structures, reducing anisotropy [11]. MD was consistently higher in the outer ring, reflecting the accumulation of oligodendrocyte precursors, immature interneurons, and progenitors in the cortical-plate-like region during months 4–5 [11].MD increased with diffusion time (Δ), consistent with CEXI model predictions of water exchange between cell bodies and the extracellular space. Conversely, MK decreased with Δ, as higher permeability and longer diffusion times blur compartmental distinctions, leading to more homogeneous diffusion and reduced kurtosis. Time-dependent diffusion MRI sensitively captures microstructure in human cortical organoids. The diffusion time-dependent increase in mean diffusivity, supported by CEXI model simulations, indicates active water exchange between compartments. This exchange-driven MD pattern offers a non-invasive marker of early cellular development, highlighting the value of dMRI and organoid models in studying human cortical formation.
Andrés LE BOEUF FLÓ (Lausanne, Switzerland), Jonathan RAFAEL-PATIÑO, Ekin TASKIN, Theo RIBIERRE, Jean-Philippe THIRAN, Erick Jorge CANALES-RODRÍGUEZ, Elda FISCHI-GOMEZ
13:30 - 15:00
#47729 - PG227 Non-invasive quantification of myelin sheath radius: Towards a diffusion MRI model for random walks confined to cylindrical surfaces.
PG227 Non-invasive quantification of myelin sheath radius: Towards a diffusion MRI model for random walks confined to cylindrical surfaces.
Quantifying the myelin sheath radius of myelinated axons in vivo is important for understanding, diagnosing, and monitoring various neurological disorders. Despite advancements in diffusion MRI (dMRI) microstructure techniques, models specifically designed to estimate myelin sheath radii remain unavailable. Recent studies suggest that it is possible to acquire dMRI data significantly weighted by myelin water using T1-based double inversion recovery (DIR) and T2-based relaxation selective measurements with short echo times (TE). Thus, it is the right moment to develop new microstructure dMRI models for this habitually neglected white matter (WM) compartment.
In this proof-of-concept theoretical study, we propose two novel dMRI models that characterize the signal from water diffusion confined to cylindrical surfaces, approximating myelin water diffusion (1). In the first, more general model, we derived an exact analytical expression for the dMRI signal using the diffusion propagator formalism based on the narrow pulse approximation. The second model employs a Gaussian approximation with time-dependent radial diffusivity, offering a simpler analytical relationship. We also developed approximate signal expressions for PGSE protocols with trapezoidal and rectangular diffusion gradients, extending beyond the narrow pulse assumption. We derive the spherical mean signals for the proposed models, which conveniently eliminate fiber orientation and dispersion effects. For the model based on the Gaussian approximation, the radial diffusivity depends on the cylinder radius r as, D⊥ = (r2/2t) [1-exp(-Dt/r2)], where t=Δ+δ is the total diffusion encoding time of the PGSE sequence and D is the axial diffusivity parallel to the main cylinder axis. These models are further extended to account for multiple concentric cylinders, mimicking the layered structure of myelin. Additionally, we introduce a method to convert histological distributions of axonal inner radii from the literature into myelin sheath radius distributions and derive analytical expressions to estimate the effective myelin sheath radius expected from these distributions (e.g., using a single-cylinder model). Figure 1 shows a schematic transverse section of a myelinated axon. We approximate the diffusion process along this spiral trajectory as diffusion within a series of impermeable concentric solid cylinders separated by infinitesimal water-filled gaps. Monte Carlo (MC) simulations conducted in cylindrical and spiral geometries were carried out to validate the models employing a maximum gradient strength of G=500 mT/m. Figure 2 shows an example of the agreement among the MC-generated signals and those predicted by the general analytical model and the Gaussian approximation for different b-values and a range of cylinder radii. Additionally, we found a significant agreement between the effective myelin sheath radii estimated from reported histological distributions and the effective radius obtained by fitting a single-cylinder dMRI model to the MC signals generated from these distributions; see Figure 3 (for more details see (1)). Despite our promising results, some limitations of this study must be addressed in future work. While the analytical models closely match MC simulations for various experimental conditions, discrepancies arise at high b-values and large diffusivities. These inaccuracies are due to the assumptions behind the narrow pulse approximation, which, despite the correction framework we introduced for more general PGSE sequences, remains an approximation valid primarily for Gaussian diffusion (low b-values). All results presented in this study are based on synthetic signals derived from the proposed analytical models or MC numerical simulations. Validation with real dMRI data, including histological analyses of various brain regions, is crucial for future work. The proposed models hold the potential for estimating the effective myelin sheath radius from real dMRI data. For example, in diffusion-T1 experiments using inversion recovery sequences effectively isolating signals arising from myelin water, as outlined by (2), our models could be applied directly to fit the measured data. Similarly, for acquisition sequences where signals from other compartments are not entirely suppressed—such as in the diffusion-T2 hybrid sequences proposed by (3,4) or the magnetization-prepared dMRI sequence by (5)—our models could be integrated into a multi-compartment dMRI model to selectively fit the myelin water component. These approaches could be applied to both ex vivo and in vivo data, employing scanners equipped with strong diffusion gradients, where recent advances (6–8) make it feasible to enhance myelin water dMRI signal contribution by reducing the TE.
Erick J CANALES-RODRÍGUEZ (Lausanne, Switzerland), Chantal M.w. TAX, Elda FISCHI-GOMEZ, Derek K. JONES, Jean-Philippe THIRAN, Jonathan Rafael PATINO
13:30 - 15:00
#47637 - PG228 Open-source Liver T1 mapping sequence - full coverage in one breath hold with 2D SMS-MP2RAGE and optimized interleaved delays.
PG228 Open-source Liver T1 mapping sequence - full coverage in one breath hold with 2D SMS-MP2RAGE and optimized interleaved delays.
Liver T1 mapping is a helpful MRI technique to depict diseases like fibrosis or cirrhosis [1]. Whole-liver T1 mapping has been performed with 3D Variable Flip Angle sequences but may exhibit higher bias and lower repeatability/reproducibility compared to 2D approaches, like Look-Locker [1,2].
However, current 2D T1 mapping methods are limited to the acquisition of only 1 to 3 slices in one breath-hold.
One way to circumvent this problem is the use of the Simultaneous Multislice (SMS) method where several slices are acquired at the same time and unfolded during the reconstruction using the sensitivity coils information. This method has successfully been used for cardiac T1 mapping [3]
The MP2RAGE sequence is another method known for providing reliable T1 maps. A 2D multislice version of the MP2RAGE has previously been proposed for rapid T1 mapping in mice [4]. Six slices could be acquired in only 9 seconds by cleverly using “dead times” of the sequence to interleave the inversion and readout of the other slices.
This abstract describes how we brings all these ideas together using open-source tools for sequence development (Pulseq [5] and KomaMRI.jl [6] ) in order to develop a SMS 2D-MP2RAGE and achieve a rapid full-coverage liver T1 mapping in a single breath-hold on a 3T siemens.
A Multi-Slice MP2RAGE sequence previously developed on a Bruker 7T scanner [4] was transferred to a 3T Siemens human scanner and modified to include SMS pulses and calibration lines (Fig 1).
For sequence development, simulation and validation of the developed sequence, we used Pulseq and KomaMRI tools. SMS excitation (sinc shape) and inversion (secant hyperbolic shape) RF pulses were generated by the sum of 2 RF shapes with an additional temporal phase term. Additional gradient echo calibration lines (24 for each slice) were added at the end of the MP2RAGE acquisition.
The main parameters of the sequence were : TI1/TI2 = 1000/3000ms, flip angle TI1/TI2 = 7°/7°, TR = 5ms, Echo Train Length = 128, SMS Slices = 6 (Total unfolded slices = 12), Slice thickness = 8 mm, Slice gap = 4 mm, matrix = 128 x 128, FOV = 320x320x144mm, in-plane spatial resolution = 2.5 mm x 2.5 mm, total acquisition time = 8.66 (MP2RAGE) + 1.44 (calibration) = 10.1 seconds.
A direct SMS reconstruction was applied using the coil sensitivity maps computed from the calibration lines using the espirit algorithm from MRIReco.jl [7] then the quantitative maps were generated using a lookup table calculated without steady-states. Pulseq sequence format can be used for both the acquisition as well as for simulation with KomaMRI.jl. This feature is especially useful to validate that the sequence and reconstruction algorithm are working before planning any experiments.
Simulations with KomaMRI were used to: First, validate the slice profile selectivity of SMS excitation and inversion pulses. The second simulation on a numerical brain phantom validates that we were able to generate the FOV/2 offset of one of the slices.
Of note, simulation helps to debug a lot of aspects of a sequence but requires well defined simulation parameters. For example, in Figure 2 first row, the” slice 2” was not the same one as on the SMS images. After investigation, it was an issue with the simulation rf timestep parameter which was too high to correctly capture the linear phase necessary to generate the slice offset, and results in aliasing of the slice excitation. Decreasing the timestep from 50us to 25us solved the issue (Fig 2 second row).
After the success of all the simulations, the acquisition was performed on a healthy subject during a breath-hold. The T1 maps, covering the whole liver, were reconstructed after unfolding the slices and (Fig 4). The mean liver T1 values (781ms +/- 50 ms) was consistent with literature.
Before applying the newly-developed sequence on humans, Figu 2 third row shows the unfolding of the SMS acquisition and validates that the reconstruction algorithm is working as expected. Using an Open-Source framework for sequence development is particularly useful when the access to a scanner is limited like in a clinical environment. The implementation of a sequence sufficiently well described in a publication can be done in a few days. Simulation is crucial to validate each key step, like the SMS RF pulses in our case.
The next steps will consist in validating the SMS-MP2RAGE sequence against gold-standard methods and investigate sensitivity to motion, B1 and B0 field inhomogeneities.
In order to further accelerate the method without changing the echo train length, parallel imaging will be implemented and the number of calibration lines will be optimized to keep the total scan acquisition as low as possible. In this work, we demonstrated the feasibility of developing a rapid, full-coverage liver T1 mapping sequence using open-source tools. These tools facilitated in this work the enhancement of a standard MP2RAGE sequence with advanced acceleration methods like SMS.
Aurélien J. TROTIER (BORDEAUX), Nadège CORBIN, Emeline RIBOT
13:30 - 15:00
#47691 - PG229 Wideband Brain MR Elastography.
PG229 Wideband Brain MR Elastography.
Magnetic Resonance Elastography (MRE) is a non-invasive imaging technique that measures the biomechanical properties of in vivo brain tissue [1]. Brain stiffness, a potential diagnostic biomarker, decreases with age and prematurely in neurodegenerative diseases [2]. However, brain MRE typically uses a narrow mechanical frequency range from 30 to 50 Hz, limiting viscoelastic dispersion analysis. A wideband MRE approach with a broader frequency spectrum aids multi-frequency dispersion analysis and potentially enhances our understanding of age- and disease-related microstructural tissue changes associated with biomechanical property changes.
This observational study recruited 15 young adults (mean age: 29.7 ± 4.1 years) and 10 older adults (mean age: 56.5 ± 3.8 years). A new wideband MRE setup was developed, incorporating two actuator designs: a cushion actuator positioned laterally beside the head for low frequencies (5-20 Hz) and a plate actuator [3] positioned beneath the head for higher frequencies (20-50 Hz). Both actuator systems can be simultaneously fitted into a standard MR head coil, enabling continuous shear wave induction across 5-50 Hz. MRE data were acquired using a spin-echo echo-planar imaging (SE-EPI) sequence with flow-compensated motion-encoding gradients (FOV=220x220x18 mm³, Matrix=112x140x15, TE=56 ms, TR=1400 ms). Shear wave speed (SWS) maps were processed using the kMDEV inversion algorithm [3]. A frequency-SWS dispersion analysis was performed by fitting a spring-pot rheological model to SWS data to assess age-related differences in brain tissue stiffness. The wideband MRE setup successfully induced shear waves from 5 to 50 Hz. Frequency-resolved wave fields and shear wave speed (SWS) maps are shown in Figure 1. Frequency-averaged brain stiffness, measured by SWS, was lower in the older group (1.16 ± 0.03 m/s) than in the younger group (1.22 ± 0.04 m/s; p<0.001). SWS decreased linearly with age (SWS = 1.28 - 0.0021×age; R²=0.365, p=0.002), equivalent to a -0.20 % annual reduction, as shown in Figure 2. Spring-pot modeling on frequency-resolved stiffness maps showed that the stiffness modulus E decreased with age (R²=0.314, p=0.005), while the fractional exponent α increased (R²=0.236, p=0.019), as depicted in Figure 3. The dual-actuator system effectively extended the frequency range of in vivo MRE to an unprecedented wide range of more than three octaves, enabling comprehensive multi-frequency dispersion analysis. Our wideband MRE approach provides deeper insights into age-related biomechanical changes of neural tissue compared to standard mono-frequency MRE. The observed age-related decrease in SWS and changes in spring-pot parameters suggest sensitivity to microstructural tissue degradation such as the loss of neural and vascular integrity, which is known to be associated with physiological aging. Although this study validated the technical implementation of wideband MRE, future research will explore its application in patient cohorts and interventional studies. Wideband brain MRE, based on a dual-actuator design, robustly assesses brain mechanical properties in a wide frequency range from 5 to 50 Hz. Age-related differences in SWS and viscoelastic properties highlight the potential of the new MRE technique to detect microstructural changes, offering a valuable tool for studying brain aging and neurodegenerative diseases.
Jakob SCHATTENFROH (Berlin, Germany), Jan BIELING, Tom MEYER, Guillaume FLÉ, Steffen GÖRNER, Helge HERTHUM, Stefan HETZER, Ingolf SACK
13:30 - 15:00
#47932 - PG230 Blood-brain barrier water exchange in relation to amyloid, cognition and cerebrovascular burden.
PG230 Blood-brain barrier water exchange in relation to amyloid, cognition and cerebrovascular burden.
Blood-brain barrier (BBB) water exchange may serve as an early biomarker for Alzheimer's Disease (AD). Novel arterial spin labeling (ASL)-based BBB MRI sequences have the potential to become early non-invasive imaging biomarkers, especially because blood water as endogenous tracer may be sensitive to earlier and more subtle BBB changes compared to other contrast-based BBB imaging techniques. Novel multi-echo BBB-ASL sequences have already been shown to be reproducible and to be associated both with aging and AD in both animal models and humans. With the recent advent of larger BBB-ASL cohorts, it becomes possible to investigate to what extent BBB-ASL is associated with aging-related vascular pathology and AD-specific pathology. This study investigated the potential of a non-invasive multi-echo arterial spin labeling-based technique to assess BBB water exchange in relation to amyloid, cognition, and cerebrovascular burden stages.
Participants older than 50 years were selected from the Center for Lifespan Changes in Brain and Cognition (LCBC) and the Dementia Disease Initiation (DDI) cohorts. All participants were scanned on the same 3T Siemens Prisma scanner with a 32-channel head coil. Two multi-post-labeling delay (PLD) Hadamard-encoded (HAD) 3D GRASE pCASL sequences were used to estimate Tex and CBF: 1) single-echo time (TE) HAD-8 with a sub-bolus duration 400 ms, PLD [600:400:3400] ms, TE = 12.5 ms; 2) multi-TE HAD-4 with sub-bolus of 1000 ms, PLD [1500:1000:3500] ms, and 8 TEs [14.4:28.9:217.2] ms. Images were analyzed with ExploreASL 1.12.0beta; CBF, Tex and ATT were quantified with FSL-FABBER and assessed in the total gray matter (GM; partial volume>0.7). Amyloid status was defined as positive (A+) or negative (A-) from the CSF amyloid-beta 42/40 ratio (cut-off ≤ 0.077) or amyloid-PET by visual reads when available. Total WMH volume was quantified with Fazekas scores (0-3) by trained neuoradiologists. Associations with age, sex, amyloid positivity (A+ or A-), cognition, and cerebrovascular burden were examined. Regarding cognition, participants were categorized into three groups: cognitively normal (CN), subjective cognitive decline (SCD), and mild cognitive impairment (MCI) (Table 1). Tex was lower in SCD and MCI compared to CN (Figure 1D), while CBF differed only between CN and MCI (Figure 1E). Tex also decreased with increasing Fazekas scores (1–2), while ATT increased in Fazekas 3 (Figure 1G-I). No association with amyloid positivity was found after age and sex adjustment (data not shown). When including all variables in the same model (Table 2, rightmost column) — age, sex, amyloid status, cognitive staging, and Fazekas scores—Tex remained significantly different between SCD and CN and between MCI and CN. In this model, CBF again showed significant differences between MCI and CN. Finally, ATT showed a significant increase in Fazekas 3 compared to Fazekas 0. Our results indicate that CBF and Tex were associated with amyloid, cognition, and cerebrovascular burden stages, though amyloid correlations did not remain significant after adjusting for all covariates. Specifically, BBB water exchange, measured by Tex, is associated with cognitive staging and with Fazekas scores and seems to be changing with these biomarkers earlier than CBF and ATT. Whether BBB water exchange alterations were the cause or effect of cognition and cerebrovascular burden cannot be differentiated from these data. In conclusion, our findings suggest that the observed BBB water exchange effects are not directly related to AD-specific pathology but seem to reflect more general aging-related neurodegenerative processes, including cerebrovascular damage. Investigating BBB water exchange across a broader spectrum of diseases and comparing multi-modal BBB imaging approaches to assess BBB dynamics, could elucidate its potential as a universal or disease-specific biomarker. Finally, these results underline the importance of assessing early cerebral microvascular pathology in aging and neurodegenerative diseases.
Beatriz ESTEVES PADRELA (Amsterdam, The Netherlands), Sandra TECELÃO, Bjørn-Eivind KIRSEBOM, Oliver GEIER, Markus H. SNEVE, Maksim SLIVKA, Amnah MAHROO, Klaus EICKEL, Matthias GÜNTHER, Kristine B. WALHOVD, Anders M. FJELL, Frederik BARKHOF, Jan PETR, Tormod FLADBY, Henk J.m.m. MUTSAERTS
13:30 - 15:00
#46011 - PG231 Placental blood-flow velocity quantification from diffusion MRI.
PG231 Placental blood-flow velocity quantification from diffusion MRI.
The placenta is vital for the long-term health of both fetus and mother[1]. Placental blood flow is associated with adverse outcomes such as preeclampsia and fetal growth restriction[2]. However, the capillary structure and blood flow characteristics in placental remain poorly understood[3]. While recent methods, such as separating maternal and fetal blood flows[4] and using T2* information[5] to extend the intravoxel incoherent motion (IVIM) model[6, 7], can capture additional information, these approaches cannot directly measure incoherent flow in capillaries. Other imaging techniques like Doppler ultrasound[8, 9] or MRI-based phase contrast angiography(PCA-MRI)[10] are either insensitive to microvascular flow or best suited for coherent flow patterns, thus, leaving a gap that this study aims to address.
We introduce a novel approach to estimate capillary-scale blood velocities using simulated diffusion MRI(dMRI) data and machine learning(ML) directly from in-vivo dMRI data, essentially adapting microstructure fingerprinting[11-13] for IVIM-type models. We construct in-silico capillary structures, simulate flow, generate dMRI signals and finally train ML regressors to learn flow parameters. Our approach outperforms the baseline IVIM, offering deeper capillary flow insights.
We present a Monte Carlo simulator for dMRI signal generation from in-silico placental capillaries, then describe velocity estimation using ML and IVIM.
Simulation
Two geometries mimicked random isotropically-orientated placental capillaries(Fig. 1): (1) a 3D ring-ball structure of three circular channels, and (2) randomly oriented cylinders, enabling simulation of IVIM-like pseudo-diffusion[6, 7]. Monte Carlo simulations tracked spin motion under flow (following capillary centerline) and diffusion (randomly in 3D), with displacement[14, 15]:x(t)=√(6D∙dt)+v∙dt, where D is the diffusion coefficient, dt the time step, and v the local spin velocity. Outside capillaries, only diffusion was considered. Internal(S_i) and external(S_e) signals were computed separately. The final synthetic dMRI signal was: :S_sythetic=V_f∙S_i+(1-V_f)∙S_e, where V_f is the perfusion fraction[16].
Key ring-ball parameters: L=[10,80]µm (vessel length), d=[5,30]µm (diameter), D =[0.001,0.003]mm²/s, v=[0.001,1]mm/s, sampled via Latin Hypercube Sampling(1000 combinations). Random cylinders considered infinite length to prevent spin trapping with d, D, and v evenly distributed (10 samples within above ranges, yielding 1000 combinations). Noise (SNR = 20) replicated in-vivo conditions[17]. These combinations generated 1000 in-silica training dMRI signals per geometry. The test dataset had 60 velocity-varying combinations(L=100 µm, d=6, 10, 14µm, D=0.002mm²/s).
Parameter Estimation
We IVIM-fitted testing dataset. S_0 and D_initial came from monoexponential fitting of b>200 s/mm^2[18]. Velocities were derived from IVIM using the Einstein relation (D^*=Lv/6)[19]. Initial V_f, L and v used the simulation parameters(ground truth), unfairly advantaging the IVIM method. Two ML regressors, Random Forest(RF) (1000 estimators, max depth 40) and Multi-layer Perceptron (MLP)(layers: 64, 32, 16, 4; learning rate 0.005) in scikit-learn, were trained on synthetic data to predict L, d, D, and v. All parameters trained jointly. Trained regressors were applied to in-vivo scans (78 cases, 69 pregnancies, from publication[20, 21]) to predict v and D maps. Fig. 2 shows capillary velocity predictions on the synthetic test dataset using RF, MLP, and IVIM, with/without noise. Fig. 3 shows predictions on in-vivo data from regressors trained on ring-ball structure only and both structures together. Fig. 4 displays velocity/diffusion coefficient trends during GA via MLP/RF. From Fig. 2, ML regressors outperformed IVIM in predicting velocities. RF and MLP showed high accuracy in noiseless data(MLP: r≈0.99; RF: r≈0.98), while IVIM performed poorly(ring-ball: r≈0.84; random cylinder: r≈0.7). With noise, MLs remained robust(MLP: r≈0.94; RF: r≈0.93), whereas IVIM worsened (r<0.55). MLs showed limitations at extremes but surpassed IVIM, which oversimplifies flow into a single pseudo-diffusion term.
Figs 3 showed training data diversity affected predictions, especially for MLP. Predicted velocities in Fig. 4 (0.3–0.9 mm/s) matched PCA-MRI findings(~0.6 mm/s)[10]. Both ML models showed a gestational decline in capillary velocity and diffusion coefficients, aligning with previous reports[22-24]. This study introduces a Monte Carlo and ML-based method for estimating placental capillary velocities, providing reliable predictions despite noise. Including more diverse structures enhances feature extraction, especially for MLP. Future work will refine the approach with more complex structures and validate using in-vivo PCA-MRI or ex-vivo perfusion imaging. Once validated, this method could non-invasively assess blood flow dynamics across organs in physiological and pathological conditions.
Zhuangjian YANG (London, United Kingdom), D.m. Cruz De OLIVEIRA, Leevi KERKELÄ, Marco PALOMBO, Elizabeth POWELL, Christopher S PARKER, Daniel CROMB, Lisa STORY, Serena COUNSELL, Kelly PAYETTE, Joseph V. HAJNAL, Hutter HUTTER, Rebecca J SHIPLEY, Daniel C ALEXANDER, Paddy J SLATOR
13:30 - 15:00
#47671 - PG232 Characterizing Water Exchange in Gliomas Using Diffusion MRI with Free Gradient Waveforms.
PG232 Characterizing Water Exchange in Gliomas Using Diffusion MRI with Free Gradient Waveforms.
Gliomas are common and aggressive tumours of the central nervous system, with a five-year survival rate of only 5% (1,2). Aquaporin 4 (AQP4) plays an important role in glioma pathology and could serve as a prognostic biomarker (3–5). AQP4 is difficult to measure directly in vivo, but can be mapped indirectly with MRI via the water exchange rate (3). However, this demands diffusion MRI (dMRI) data acquired with non-conventional approaches such as free gradient waveforms (6). In this study, we evaluate whether dMRI reveals exchange-driven contrast in gliomas that complements standard imaging parameters such as apparent diffusion coefficient (ADC), kurtosis, and contrast enhancement.
Theory:
We use the restriction-exchange framework (6,7) to analyse the MR signal, S, via
ln(S/S_0)=-b⋅E_D+1/6 b^2 E_D^2 K_T⋅(1-kΓ) #(1)
where there are four microstructural parameters: S_0 (signal without diffusion weighting), E_D (mean diffusivity), K_T (kurtosis), k (water exchange rate) and two experimental parameters: b (b-value) and Γ (exchange-weighting) given by
Γ=2/b^2 ∫_0^T t q^2 (t) q^2 (t+t' )dt' #(2)
where q(t)=γ∫_0^t g(t') dt' is the q-vector and γ is the gyromagnetic ratio. Waveforms are designed with varying Γ but fixed sensitivity to restricted diffusion to enable selective sensitivity to exchange.
Gradient waveform design:
Two exchange-encoding gradient waveforms were optimised following (6) (Fig. 1A). Monte Carlo simulations were used to verify the exchange weighting properties of the waveforms (Fig. 1B).
Participants:
In this exploratory study, five participants with primary brain tumours (cases 1 through 5) aged between 56 and 74 were examined. Cases 1-4 were diagnosed with grade 4 glioblastoma and case 5 with astrocytoma.
MRI scans:
All participants were scanned on a 3 T MAGNETOM Prisma (Siemens Healthineers, Forchheim, Germany) using a research sequence (8). The diffusion imaging used b-values of 0, 1.3, 2.6, and 4 ms/µm2 in 10 directions with a spatial resolution of 2×2×5 mm3, TE = 138 ms, TR = 4.6 s, strong fat suppression and acquisition time of 5 min. One participant was scanned longitudinally at 4 timepoints.
Data analysis:
Acquired images were denoised (9), motion- and eddy-current corrected (10), and powder-averaged. Exchange-driven signal contrast was calculated using
ΔS(b) = ((S_(low Γ) (b) - S_(high Γ) (b)))/(1/2 (S_(low Γ) (b)+S_(high Γ) (b)) ) #(3)
where S_(low Γ) (b) and S_(high Γ) (b) are signals acquired with waveforms at Γ=10 and Γ=40 ms, respectively, at a fixed b-value. ResEx parameter estimates were obtained by fitting Eq. (1). Explorative ROI-based analyses were performed using BraTS (11) to compare exchange estimates in diseased and healthy tissues. Given the small cohort, results were interpreted qualitatively without formal statistical testing. Figure 1 illustrates that the two waveforms are specific to exchange.
Figure 2 shows that exchange contrasts manifest in gliomas at higher b-values, as expected since exchange is a higher-order time-dependent effect. The contrasts show different trends across cases. Gd enhancement is connected to increased exchange contrast. ADC is high in oedema and necrotic regions where cellularity is likely low. Kurtosis maps show opposite patterns. Exchange maps resemble the contrast patterns.
Fig. 3 shows exchange estimates (k) in oedema compared with contralateral healthy white matter. Some cases show slight elevation in k in the oedema, others a marked increase, suggesting disrupted membrane integrity.
Figure 4 shows T1-weighted images highlighting progressive enlargement of both the core and enhancing tumour over time. ADC is consistently high in the core, and kurtosis is high in the enhancing rim. Of note, exchange hyperintensity extends beyond the enhancing rim over time, indicating tumour infiltration into peritumoural tissue. Elevated water exchange rates, as measured by dMRI with free gradient waveforms, were detected in all five glioma patients. Exchange maps revealed tumour heterogeneity not captured by ADC or Gd enhancement, indicating potential for non-invasive assessment of membrane properties. Elevated exchange in tumours may reflect increased membrane permeability, possibly due to upregulation of AQP4 (12) or disruption of the blood-brain barrier (13) although the latter is an unlikely explanation due to the high b-values we used. These findings have both methodological and clinical implications, since accounting for exchange improves kurtosis estimates, which have been shown to aid in glioma grading (14,15). While limited by a small sample size, this study provides a proof-of-concept for in vivo mapping of water exchange in gliomas. In conclusion, dMRI allows non-invasive mapping of exchange in gliomas and adds information to standard imaging. Exchange is related to AQP4 expression and is thus a potential biomarker of tumour aggressiveness. Future studies with larger cohorts are warranted to evaluate the diagnostic utility of exchange imaging.
Arthur CHAKWIZIRA (Lund, Sweden), Filip SZCZEPANKIEWICZ, Carl-Fredrik WESTIN, Linda KNUTSSON, Pia SUNDGREN, Markus NILSSON
13:30 - 15:00
#47749 - PG233 Towards B0 and B1 robust spiral turbo spin echo sequence at 7T.
PG233 Towards B0 and B1 robust spiral turbo spin echo sequence at 7T.
Turbo Spin Echo (TSE) imaging at 7T faces increased challenges due to shortened T2 relaxation times at 7T restrict the echo train length (ETL). At the same time, longer T1 relaxation times require longer repetition times prolonging time per shot. Spiral TSE sequences, which combine high signal acquisition efficiency with reduced number of shots and shorter ETLs, are therefore particularly attractive at 7T for achieving high-resolution imaging within shot scan times.
However, higher field strengths bring the challenge of inhomogeneous transmit (B1+) fields. Especially TSE sequences suffer from it, which impair uniform excitation and refocusing and designing slice-selective pTx pulses is complex. Moreover, spiral readouts will be affected by B0 inhomogeneities, resulting in a more complex k-space filter as in Cartesian sampling.
To address these challenges, we propose a 7T spiral TSE sequence based on the 3T spiral TSE sequence by Hennig et al. [1], incorporating shorter spiral segments to reduce B0 sensitivity and applying Direct Encoded Signal Control (DESC) [2] to mitigate B1+ inhomogeneities. DESC allows the use of standard slice selective RF pulses while optimizing the shim mode for each pulse individually.
We employed a spiral TSE sequence [1] (encoding scheme in Fig. 1) in fixed mode with a fat saturation pulse at the beginning with a fixed cp shim mode. The sequence was implemented with a short echo spacing of 6 ms to minimize off-resonance effects, an effective echo time of 12 ms and a total acquisition time of 0.42 s. The ETL is 68, including one dummy pulse. A 90° excitation is followed by a 40° dummy refocusing pulse and subsequent 40° refocusing pulses. Except for the fat saturation pulse (cp mode), all shim modes were optimized individually.
For B1-shim optimization, we used the MR-zero [3] framework with a differentiable Phase Distribution Graph [4] simulation written in PyTorch, enabling gradient-based optimization [5,6]. The loss function is the RMS of the magnitude difference between the reconstructed and ideal target image, which is simulated with constant, mean relative B1+ value of the cp mode. The simulation phantom used a combination of BrainWeb [7] data parameterized with reasonable relaxation values and multi-channel B1+ maps and a B0 map measured at 7T using the DREAM approach [8,9]. A low-resolution (42×42) phantom and static shim initialization were used to accelerate optimization. We optimized the B1-shims for the first 17 pulses (1 excitation pulse, one dummy refocusing pulse, 15 refocusing pulses); subsequent pulses used static shim mode. The resulting shim modes were exported using the pTx extension of the Pulseq standard [10,11]. Image reconstruction was performed at a resolution of 256x256 using NUFFT with and without off-resonance correction to address for B0 influence during the spiral TSE acquisition.
The measurements were performed on a 7T MAGNETOM Terra.X scanner (Siemens Healthcare, Erlangen) on one healthy subject after written informed consent and approved by the local ethics committee. The B1-shim optimization took approximately 3 minutes for 100 iterations and results in different shim magnitudes and phases for each optimized pulse. The resulting pulse-specific B1+ fields are illustrated in Fig. 2 (transversal orientation) and Fig. 3 (sagittal orientation). Notably, the excitation pulse achieves a higher mean B1+ factor compared to both cp mode or static shimming. The final image exhibited improved homogeneity relative to static shimming method (Fig. 4). Receive fields are eliminated. Despite improvements, residual image artifacts remain visible in both cases. It was shown the first spiral TSE MR image at 7T with a long ETL, effectively limiting the impact of B0 inhomogeneities, additional off-resonance correction at NUFFT reconstruction stage had minor influence on the image quality. By performing a DESC optimization, we optimized the shim parameters of individual RF pulses within the sequence leading to improved image homogeneity. The duration of the optimization process could be further reduced by selecting a smarter initial guess and/or utilizing idle scanner time for measuring other clinical sequences in parallel.
Despite this improvement, residual image artifacts remain present. These could result from flow effects, or gradient imperfection and will be investigated in the future.
The minimal effect of the off-resonance correction indicates the potential to use longer echo spacing, which will allow larger spiral segments per ETL and thus will lead to reduction of the overall echo train length. We demonstrated the first spiral TSE MR image at 7T effectively limiting the impact of B0 and B1 inhomogeneities.
Simon WEINMÜLLER (Erlangen, Germany), Jürgen HENNIG, Felix DIETZ, Peter DAWOOD, Jonathan ENDRES, Moritz ZAISS
13:30 - 15:00
#47716 - PG234 Towards faster myelin bilayer mapping: Reducing the specific absorption rate at no cost.
PG234 Towards faster myelin bilayer mapping: Reducing the specific absorption rate at no cost.
Myelin bilayer mapping is a recently developed method for quantitative mapping of macromolecular myelin content in the brain. Signals stemming directly from the myelin lipid-protein bilayer are captured at multiple TEs using dedicated short-T2 hardware[1-3] and the zero echo time (ZTE)[4] sequence variant with hybrid filling (HYFI)[5], and subsequently separated from other signal components via a fitting procedure[6].
A key feature of ZTE sequences is that the gradient is ramped up before RF excitation. To excite the full FOV, the RF pulse bandwidth (BW) must therefore match the image BW. Short-T2 imaging requires rapid spatial encoding[7], which is achieved through strong gradients, yielding high image BWs. Consequently, high-BW excitation pulses are needed, which have high specific absorption rates (SAR). This high SAR limits the achievable flip angle (FA) to below the Ernst angle, meaning that the SNR is SAR limited. Low SNR, in turn, is compensated by increased scan time.
The multi-TE protocol is designed such that shorter TEs have higher image BWs. So far, the same frequency-swept[8] RF pulse, with BW matching that of the TEmin image, has been used across all TEs for consistent excitation. As such, every image has reached the SAR limit, SARmax. For frequency-swept pulses, FA ∝ 1/sqrt(pulse BW), i.e., lower pulse BW gives a higher FA, so lower RF power is needed to reach the same FA. Therefore, by instead matching the pulse BW to the respective image BW, SAR is reduced for longer-TE images.
To benefit from this reduction, the SAR safety limits must be considered. These are defined as moving time averages[9]: SAR ≤ SARmax per 6 min window, and SAR ≤ 2⨯SARmax per 10 s window. Reducing SAR in terms of the safety limits thus boils down to decreasing the limiting average SAR, denoted SAR*, which can be realised by efficient distribution of the protocol SAR.
Here, we apply this concept to myelin bilayer mapping by optimising the RF pulses for each TE, splitting the images into shorter scans, and interleaving the scans to minimise SAR*.
A protocol with eight TEs is used; scan parameters are listed in Table 1. The RF pulses were chosen through an optimisation procedure to match the FA of the reference pulse (i.e., that of the TEmin image) at minimal RF power.
For the interleaving, each signal average is split into three scans of 70–85 s each (depending on the angular undersampling factor). Each scan needs 5 s of setup time (no SAR) and 3 s of dummy excitations (with SAR), leading to a total scan time of 53:31 without splitting and 57:40 with splitting. The scans are executed in an order that minimises SAR*, as determined by a custom algorithm that tries to reach the optimal SAR* reduction (i.e., that achieved if the total protocol SAR is spread evenly over the full scan time): It places one scan at a time (first trying those with highest SAR) and checks if the optimal reduction is met. If no scan can be placed, it lowers the expected reduction and tries again.
Each set of three scans is reconstructed into an image, the images are registered to each other, and signal averages of the same image are combined. Otherwise, the processing is the same as previously[6], including k-space filtering and extension, bias correction, and model fitting to produce the quantitative myelin bilayer map.
A healthy 57-year-old male volunteer was scanned over two sessions, once with interleaving of the protocol and once without. The relative SAR of the protocol without and with interleaving is shown in Figures 1 and 2, respectively. It is clear that the 6 min window represents the limiting average. With interleaving, the SAR* is reduced by 44.3%, which, when adjusted for the increased scan time, corresponds to an effective reduction of 39.9%.
Myelin maps for the data with and without interleaving are shown in Figure 3 for similar slices. The map quality is slightly worse for the data with interleaving, but this is expected to be solved with due fine-tuning on the acquisition side. Assuming a linear relation between FA and SNR, the achieved reduction in SAR* could be used to increase the SNR by ~40%, which could, ideally, be traded for a ~44% reduction in scan time, i.e., a total scan time of ~32 mins.
The choice of splitting factor is flexible, and can be set individually per image. It represents a trade-off between widening the optimisation space and increasing the scan time (due to larger overhead). The uniform factor of three used here was chosen for demonstration purposes and is not necessarily optimal. Similarly, the scan setup duration can essentially be set freely (above a lower limit). As such, there is still room for further improvements. Myelin bilayer mapping was successfully performed at 39.9% lower SAR* with comparable map quality. In future, this SAR* reduction could be traded for increased SNR or reduced scan time. Similar SAR* reduction options may be available on clinical systems depending on the SAR-monitoring implementation.
Emily Louise BAADSVIK (Zurich, Switzerland), Markus WEIGER, Ariel Martinez SILBERSTEIN, Roger LUECHINGER, Klaas Paul PRUESSMANN
13:30 - 15:00
#47319 - PG235 Exploring the potential of non-invasive perfusion imaging in the liver using velocity-selective Arterial Spin Labeling by comparing different labeling schemes.
PG235 Exploring the potential of non-invasive perfusion imaging in the liver using velocity-selective Arterial Spin Labeling by comparing different labeling schemes.
Liver diseases are prevalent, and for their identification and characterization, imaging perfusion is essential [1]. Arterial Spin Labeling (ASL) [2] is a non-invasive technique used for measuring perfusion without the need for contrast agents, typically by magnetically labeling the blood flowing into the organ of interest. Velocity Selective ASL (VSASL) [3] is a specialized method that labels blood based on its velocity directly in the imaging region. This method is advantageous since it eliminates the need for additional time for blood to reach the imaging region. Moreover, due to the challenge of dual blood supply from the hepatic artery and portal vein, this method is especially beneficial for liver imaging [4]. In this preliminary study, three different VSASL preparation schemes are compared and the quantified perfusion values determined.
For the determination of the perfusion in the liver, an in-house developed velocity selective saturation sequence (Gaussian compRefoc) [5], a velocity selective inversion (VSI) [6] and a VSI sequence with sinc modulation of the nine RF pulses (VSI sincMod) [7] were compared. The images were acquired under time-controlled breathing to minimize motion-induced artifacts on a 3T scanner (vidaFit, Siemens Healthineers, Erlangen, Germany) using the vendor independent sequence development framework gammaSTAR [8]. For the quantification of the hepatic blood flow (HBF), the general kinetic model for VSASL [9] was used and a mean value over the whole liver determined. For a test-retest regarding the robustness of the sequence, the Gaussian compRefoc was tested in the same healthy volunteer (30y, female) in two additional exams.
Following parameters were used: TR=6500ms, TE=23ms, velo = 2 cm/s in z-direction, TI=1200ms, PLD=100ms, velocity-selective insensitive control-module, meas=12 including a phase cycling of the refocusing RF pulses for all three versions. For the M0 acquisitions the same VSASL sequences with TI=10s and no background suppression pulses were used. Figure 1 shows the calculated perfusion-weighted images (PWI) for the three different VSASL techniques in the liver. It can be observed that the Gaussian compRefoc sequence displays the most homogeneous perfusion signal throughout the liver. In comparison, perfusion signal can also be measured in the VSI and VSI sincMod sequence acquisitions, but some regions of the liver are low in signal intensity, therefore, the overall signal is not as homogeneous as in the Gaussian compRefoc sequence. The quantification of the hepatic blood flow (Table 1) shows that the quantified values are slightly lower than the global hepatic blood flow measured previously with pseudo-continuous ASL [10]. The test-retest results of the Gaussian compRefoc sequence (Figure 2) show that no homogeneous perfusion signal throughout the liver could be measured in the second measurement, and a signal progression from anterior to posterior, from positive to negative values, can be observed. However, the third measurement is comparable to the first one. The acquisitions in Figure 1 clearly demonstrate the possibility of non-invasively measuring perfusion in the liver using all three VSASL techniques. Overall, the Gaussian compRefoc sequence displays the most homogeneous perfusion-weighted signal. This could be due to the fact that this sequence is specifically designed to be insensitive to B1-field inhomogeneities, which often occur in the liver [4]. The hepatic blood flow quantified with all three VSASL techniques yields reasonable values. The slight decrease could be explained by the different strengths of labelling the arterial or venous part due to varying blood flow velocities, which leads to a representation that isn't truly global perfusion. The test-retest results of the Gaussian compRefoc sequence demonstrate the sequence’s sensitivity to confounding effects. For instance, a suboptimal shimming process could have resulted in the decreased signal inside the liver in the second measurement. In the future, it will be necessary to acquire more data and enhance the robustness of the sequence, such as implementing an advanced and quick shimming process to homogenize the field, thereby stabilizing the signal. In conclusion, the results demonstrate the ability to measure hepatic blood flow comparably using all three VSASL techniques. Among these, the specially-developed Gaussian compRefoc sequence yields the most reasonable results, assuming that the optimum of all system parameters is achieved during the preparation process. Future research aimed at stabilizing this preparation process is expected to enhance the robustness of the sequence.
Mareike Alicja BUCK (Bremen, Germany), Jörn HUBER, Daniel Christopher HOINKISS, Matthias GÜNTHER
13:30 - 15:00
#45798 - PG236 Extending the multi-parametric IR-GE-SE EPIK sequence to the simultaneous quantification of T1, T2 and T2* relaxometry.
PG236 Extending the multi-parametric IR-GE-SE EPIK sequence to the simultaneous quantification of T1, T2 and T2* relaxometry.
Our group has recently developed a novel, fast multi-contrast GE-SE sequence [1] based on EPI keyhole techniques [2] to enhance spatiotemporal resolution and the number of echo samplings by incorporating two spin echoes for improved T2 quantification. The resulting sequence offers multi-parametric output in the form of simultaneous T2, T2*, vCBV and OEF estimation. The present work aims to extend the sequence variability to offer T1 quantification based on inversion recovery sampling.
The existing 10-echo GE-SE EPIK sequence [1] was extended by including initial slab selective inversion pulses of 200mm in width. Thereby, repeated IR-GE-SE acquisitions that sample different inversion times enable T1 recovery sampling [3]. It is important to note that, due to the specific EPI with keyhole methodology [4], all slices are acquired after each inversion pulse, therefore leading to different inversion time (TI) ranges with increments of (TR/slice number). The resulting echo times are 12, 24, 44, 56, 68, 80, 92, 112, 124 and 136 ms. Phantom and in vivo datasets of 10 slices with a TR of 2s were acquired with an acquisition time of 45s per TI for a spatial resolution of 1.9x1.9x3mm3. Eleven different TIs of 25, 100, 250, 500, 750, 1000, 1250, 1500, 2000, 2500, and 3000 ms were acquired. T2 and T2* maps were computed for each TI acquired in order to test the quantification stability. The sequence diagram of the IR-GE-SE EPIK sequence is shown in Figure 1. T2 and T2* quantification based on the 10-echo data acquired at different TIs are in agreement with the literature and match the previous standard GE-SE EPIK quantification results. Exemplary maps are shown in Figure 2. The T1 maps show a good contrast quality, although the quantified values underestimated the expected values by 20-40%. The resulting T1, T2 and T2* distributions for the in vivo data sets are summarized in Figure 3a. Summarized mean values from the sphere phantom and WM/GM regions of the in vivo data are shown in Figure 4 alongside the expected values from reference acquisitions. Figure 3 (b, c) shows the resulting mean T1 value in a WM ROI for each slice to demonstrate the dependency of T1 on the actual TI range as the latter changes with the slice count. An overall increase in mean WM T1 can be seen across the slices with a plateau over the central six slices. In addition, Figure 5 shows the mean whole-brain T2 and T2* values obtained from each TI echo-series acquired. While a minor decrease in T2 is observed, T2* is robust and the quantification is stable for TI larger than 500ms overall. While the map quality and T2/T2* robustness indicate the potential of extending the 10-echo GE-SE EPIK to T1 mapping, future work is needed on correcting the T1 underestimation. Potential reasons may be insufficient signal recovery or some cross-temporal effects arising from the EPIK sparse iterations to recombine single-weighted images. The T1 preparation method could be improved by using a TAPIR-like methodology [5] or slice-shuffling technique [6]. Moreover, a simple 2-point methodology will be tested in the future. All of these approaches should be directly compared to measured gold-standard values rather than expected literature values. In terms of T2 and T2*, the repeated acquisitions of different TIs may be useful for a suitable averaging method. IR-GE-SE EPIK data offers quick access to simultanoues quantification of transverse and longitudinal relaxation times. Especially T2 and T2* show a great robustness and accuracy, while T1 contrast shows the potential towards an additional quantitative biomarker but requires future correction of its current understimation. The clinical relevance of a multi-parametric fast imaging sequence can be reached in future work by optimizing the IR-GE-SE EPIK perfomance.
N. Jon SHAH, Fabian KÜPPERS (FZ Jülich, Germany)
13:30 - 15:00
#47773 - PG237 Mapping Brain Iron with Quantitative MRI: Validation and Applications.
PG237 Mapping Brain Iron with Quantitative MRI: Validation and Applications.
Iron regulation in the brain, influenced by compounds such as ferritin, transferrin, and ferrous ions, is critical for normal brain function and is implicated in aging, neurodegenerative diseases. Recently, a quantitative MRI (qMRI) based approach was proposed to in vivo estimate the iron homeostasis [1]. In this method the dependency between two MRI relaxation R1 and R2* rate constants is estimated (r1-r2* relaxivity). In this work we validate this qMRI technology using an independent dataset of healthy young and old subjects and Parkinson’s disease (PD) patients, by comparing it to known histological results [2], [3].
qMRI scan from 87 subjects scanned in the Hebrew University [4]. From each subject R1, R2* and the r1-r2* relaxivity were estimated in 11 brain regions of interest (ROI). In vivo brain values (R1, R2*, r1-r2* relaxivity) were compared with ex vivo published quantification of iron compounds on the same ROIs. Our analysis revealed consistent results, with in vivo qMRI scans correlating with ex vivo iron quantification. Specifically, the R2* highest correlation was to iron content (R-sq=0.76 p=8.1e-9) and the r1-r2* highest correlation was with iron mobilization (R-sq=0.68 p=1.6e-7). This reinforces the reliability of this qMRI-based method for assessing brain both iron fraction and iron homeostasis. A detailed analysis using ANOVA and Tukey's post-hoc tests to compare groups within the regions of ex vivo and in vivo values revealed inconsistencies in group differentiation between iron levels and R2* in the Globus Pallidus and Putamen, as well as between r1-r2* and iron mobilization in the Globus Pallidus and Caudate. These discrepancies may be attributed to the limited number of subjects. This non-invasive qMRI method offers a valuable tool for monitoring brain iron homeostasis and could enhance the diagnosis and understanding of disorders related to impaired iron regulation. Establishing the relationship between in vivo and ex vivo iron regulation opens the way for future research relating specific brain regions at disease states (e.g., PD) and the impact of specific genetic subgroups.
Maryana POZINA (Jerusalem, Israel), Elior DRORI, Lee COHEN, Gilad YAHALOM, Aviv MEZER
13:30 - 15:00
#46623 - PG238 The MP2DESS: a new method for simultaneous T1 and T2 measurements in knee imaging.
PG238 The MP2DESS: a new method for simultaneous T1 and T2 measurements in knee imaging.
In musculoskeletal MRI, the T2 relaxation time is the most commonly estimated quantitative parameter, as it is an indicator of the extracellular matrix state. T2 mapping can serve as an important marker for therapeutic monitoring of tissue changes during disease progression or recovery [1,2] . The Dual Echo Steady State (DESS) sequence is of particular interest to perform such studies[3,4]. In addition to providing high contrast structural images, T2 maps can be obtained in short scan times. Nevertheless, two improvements are needed: 1) perform high spatial resolution in 3D without lengthening scan duration and 2) provide an accurate 3D T1 map to precisely estimate T2. To circumvent both issues, the DESS sequence was inserted into the 3D MP2RAGE sequence diagram [5], and the subsequent method, called MP2DESS, was evaluated on one volunteer at 3T.
The gradient echo encoding within the two echo trains of the MP2RAGE were replaced by a DESS encoding, enabling the reconstruction of two images per train: FID1, Echo1 and FID2, Echo2, respectively (Figure 1). The T1 and T2 maps were obtained by using two different and known T1- (MP2RAGE ratio proposed by Marques et al [5]) and T2- (DESS ratio proposed by Bruder et al [6]) sensitive image combinations to iteratively estimate each parameter, similarly to the method proposed by Heule et al [7]. A T2 map was estimated via look up table dictionary matching with the DESS ratio, which would then be used to measure a new T1 map, with a matching of the MP2RAGE ratio instead, and vice-versa. This two-way iterative estimation would then be run until convergence of the two parameters (Figure 2). Simulations demonstrated that this convergence was obtained rapidly, after 3 to 5 iterations. The signal dictionaries used for the look up tables were computed using the EPG formalism in the Sycomore toolkit [8].
Multiple sets of MP2DESS parameters were employed during scans as prior simulations had demonstrated that modifying certain parameters such as the flip angle, the inversion times and the total TR could have an important impact on the estimates. These parameters were modified with the constraint of keeping the acquisition time short (<=4 min).
Acquisitions were carried out on a Siemens 3T PRISMA scanner on the knee of one healthy volunteer, with a 1mm isotropic resolution and the following parameters: Compressed Sensing acceleration factor = 4, ETL= 120 or 150, TE1/ TE2/TR=4/7/11ms, FA =10 or 20°, MP2DESS_TR=3200, 3800 or 4300ms, acquisition time between 3min33s and 4min01s. The T1 and T2 values were compared with their respective reference methods: the MP2RAGE (1mm isotropic, TE/TR=3/7ms, FA=7°/7°, MP2RAGE_TR=5s; ETL=125)[9], the DESS (1.2mm isotropic, TE1/TE2/TR= 5/10/15ms, FA=20°)[3,10] and a B1 map. The two reference sequences were also acquired with modified parameter sets to match the MP2DESS parameters.
The relaxation times were measured in the cartilage, the meniscus and the muscle. 3D T1 and T2 maps were estimated from the reconstructed images of each MP2DESS scan. The meniscus, cartilage and muscle regions were clearly visible in both maps, in 3 out of 4 scans (MP2DESS 1,2 and 4) (Figure 3). Saturation of T1 estimates in the upper and lower regions of the images were due to low B1 values (<70%) not being included in the dictionary during the estimation. Mean T1 estimates for the three functional parameter sets were similar to the MP2RAGE reference values (<21% relative difference in the meniscus, <10% in the cartilage and <2% in the muscle, Figure 4). Mean T2 measurements in the cartilage and muscle returned <24% and <20% differences, respectively. T2 in the meniscus was estimated to be around 10ms in the MP2DESS scans, which reflects values found in literature [3], suggesting that the reference DESS scan was not optimal here (19.4ms). Modifying the parameters of the MP2RAGE and DESS sequences also impacted the value of both estimated quantities in a similar fashion. The proposed sequence and T1 and T2 estimation method allows rapid musculoskeletal scans in 3D, with accurate measurements in most regions of interest depending on the choice of parameters. Interesting next steps could include reducing the voxel size for better segmentation of the cartilage, running Monte Carlo simulations to determine optimal parameters and performing test-rests scans measurements to assess repeatability of both quantitative estimates. We have proposed a novel sequence that has the ability to measure reliable T1 and T2 estimates in the knee and benefits from rapid scan times and 3D isotropic voxel sizes.
This method could be of great interest for reproducible musculoskeletal MRI, especially for the longitudinal monitoring of cartilage repair.
Emile KADALIE, Nadège CORBIN, Aurélien TROTIER, Sylvain MIRAUX, Emeline RIBOT (Bordeaux)
13:30 - 15:00
#45996 - PG239 Defining a baseline of healthy brain function using voxel-wise rsfMRI: a pilot characterization of common metrics.
PG239 Defining a baseline of healthy brain function using voxel-wise rsfMRI: a pilot characterization of common metrics.
Resting-state functional MRI (rsfMRI) is a popular, non-invasive, indirect method to assess neuronal brain activity without specific stimuli[1,2] based on the Blood-Oxygen Level Dependent (BOLD) signal[3,4]. The BOLD signal has several features that offer unique and complementary insights into brain function and organization, which primarily focuses on functional connectivity (FC)[1]. Voxel-wise metrics such as amplitude of low-frequency fluctuations (ALFF)[5], fractional ALFF (fALFF)[6], regional homogeneity (ReHo)[7], entropy[8], and complexity[9] allow for reliable analyses of other features of the BOLD signal. However, some of these have had minimal usage in rsfMRI analysis compared to the more popular FC approaches[2,10], neglecting alternative characteristics of the BOLD signal. One problem with this, for example, is the lack of characterization of normative data to establish the localized rsfMRI baseline. Therefore, this pilot study investigated the normative range of various rsfMRI analyses in healthy subjects, establishing a foundational baseline for future research.
rsfMRI scans were retrieved from the open-access International Neuroimaging Data-sharing Initiative, available at https://www.nitrc.org/projects/fcon_1000/. rsfMRI brain scans from 50 participants (50:50 sex ratio, 20-40 years old) were used from the Nathan Kline Institute Rockland Sample. Data were collected on a 3T Siemens MAGNETOM Trio with the following parameters: resting state BOLD, Axial 2D, GE-EPI, 3mm isotropic, FOV=21.6cm, TE/TR=30/2500ms, 260 time points.
Data were preprocessed as follows: (1) deletion of the first five functional volumes for magnetization equilibrium, (2) eddy current correction, (3) motion correction, (4) spatial smoothing with Gaussian kernel and FWHM=5mm, (5) 4D global normalization, (6) brain extraction and (7) registration to MNI space. Linear detrending and temporal filtering were applied according to the prerequisites of each analysis. After preprocessing, six rsfMRI analyses were performed: ALFF and fALFF, using the FFT-based approach[5,6,11]; ReHo, utilizing Kendall's coefficient of concordance (KCC)[7,12]; entropy, employing Approximate Entropy (ApEn) based on the average uncertainty of a time series[8,13,14]; fractal complexity through the Hurst exponent (H) using the rescaled range (R/S) method[15–17]; and chaos and regularity using the Lyapunov exponent[18–20].
Voxel-wise averages of each rsfMRI analysis were computed, along with their respective whole-brain histograms, to show the regional contrast and dynamic range of each metric. In addition, descriptive statistics were computed for each rsfMRI analysis, segregating by sex and region-of-interest (ROI), as previous research has reported sex and regional differences in some of these analyses[21]. A total of six rsfMRI analyses were investigated in a sample of 50 participants. Voxel-wise whole-brain averages of each of the six metrics are shown in Fig.1, displaying different contrasts between brain regions. Histograms depicting the value distribution of each rsfMRI analysis are shown in Fig.2, where it is seen that each analysis has a different distribution, mean, and dynamic range. Fig.3 shows the median and IQR (top), and the mean and SE (bottom), both are segregated by sex (male, female) and ROI. In agreement with previous characterization, ALFF, fALFF, and ReHo voxel-wise averages showed maximal contrast between WM and GM, which were further supported by descriptive statistics[5,22]. In addition, this pilot study shows a similar behavior for H, ApEn, and Lyapunov exponent. Furthermore, the normative ranges and distributions showed distinct characteristics for each metric, with ALFF, fALFF, and ReHo having relatively higher standard deviations compared to H, ApEn, and Lyapunov exponent. Lastly, descriptive statistics suggest that ALFF, fALFF, ReHo, H, and Lyapunov exponent have higher GM values than WM, in contrast to ApEn. This pilot study aimed to provide an initial characterization and baseline of six localized rsfMRI analyses in healthy participants. Although this has been performed for some analysis in independent research[21], to the authors’ knowledge, no study has attempted to characterize and compare these rsfMRI analyses. Nevertheless, in the future, it will be necessary to increase the sample size and expand to more datasets collected at other locations. Also, we will increase sample diversity and enable the use of more powerful statistical methods, testing for significant differences in these rsfMRI analyses across sex, age[16,23], and expand the number of brain regions included. Thorough characterization of these metrics could improve understanding of the healthy brain, which could help better understand brain diseases.
A. AMADOR-TEJADA (Canada, Canada), E. DANIELLI, D.a. KUMBHARE, M.d. NOSEWORTHY
13:30 - 15:00
#47409 - PG240 Investigating the repeatability of phase-based magnetic resonance electrical properties tomography (MR-EPT) in the human brain.
PG240 Investigating the repeatability of phase-based magnetic resonance electrical properties tomography (MR-EPT) in the human brain.
Phase-based magnetic resonance electrical properties tomography (MR-EPT) is an emerging technique to non-invasively measure tissue electrical conductivity (σ) from the MR transceive phase (φ0), via the Helmholtz equation [1]. Despite technical advancements, the repeatability of EPT remains largely unstudied. In this study, we investigated the repeatability of a noise-suppressing EPT reconstruction method we developed [2], optimised and applied to the human brain.
Data acquisition:
Ten healthy volunteers (seven female, aged 23-30 years), recruited as part of a previous study [3], were scanned in two sessions one week apart, with three identical scans per session. Multi-echo 3D GRE images were acquired using a 3T Philips Achieva system. Sequence parameters are shown in Table 1A.
EPT reconstruction:
An in-house EPT pipeline [2] was applied to the denoised [4] complex GRE data. A total field map was obtained from a non-linear fit of the complex data over all TEs [5]. Residual phase wraps were removed using SEGUE [6]. The transceive phase, φ0, was estimated by predicting the complex signal at TE=0 and extrapolating the phase. Conductivity was reconstructed via the surface integral of the φ0 gradient, using 3D spherical kernels of radius 21 mm and 24 mm to perform differentiation and integration, respectively. An optimised magnitude- (Mag) and segmentation-based (Seg) weighting technique was employed, for noise suppression and edge preservation [2]. The magnitude image at the final TE was used both for Mag-weighting and to segment grey matter (GM), white matter (WM) and cerebrospinal fluid (CSF) with SynthSeg [7], for Seg-weighting. For Mag-weighting, the magnitude was adjusted by a free parameter, δ, which varied automatically based on phase noise levels [8].
Repeatability analyses:
Repeatability was assessed both within-subject (including both intra- and inter-session) and between-subject, using some of the most popular and informative repeatability metrics (listed in Table 1B) [9-11]. For regional comparisons, the median conductivity was calculated within the GM, WM and CSF, excluding non-physical negative conductivity values which were considered erroneous. To enable voxelwise comparison, each subject’s conductivity maps were co-registered using FLIRT [12, 13]. The repeatability is illustrated in Fig. 1, which shows conductivity maps in a representative subject, across all six sessions (prior to registration).
Regional variability is illustrated in Fig. 2, showing the median σ values in each tissue type, for all subjects across all six sessions. Greater variability was observed in the CSF compared to GM and WM.
All repeatability metrics are shown in Fig. 3: average normalised root mean square error (NRMSE) across repetitions, and three kinds of standard deviation (SD; within-region, within-subject, and between-subject), calculated in each tissue type. All of these metrics were significantly higher in the CSF, compared to both GM and WM (p = 0.002). The repeatability of our EPT method was reasonably high in the GM and WM regions, but was significantly poorer in the CSF. This is likely attributed to the higher rate of EPT errors in the CSF, generating negative and/or physically implausible conductivity values (> 10 S/m in the human brain). The reason for these errors remains unclear, but may relate to the increased anatomical complexity of the CSF regions, compared to more homogeneous WM areas [14].
Interestingly, in the grey and white matter, the between-subject SD was lower than the within-subject SD, suggesting that EPT is more repeatable across different subjects than within a single subject across repetitions.
Future work will aim to minimise the CSF error rate, as well as compare repeatability using different EPT reconstruction methods. In this preliminary study, we investigated the repeatability of EPT in the human brain, using a surface-integral MagSeg-weighted reconstruction method designed to suppress noise and preserve edges. Both visual inspection and quantitative analysis revealed that repeatability varies significantly between tissue types, with the poorest repeatability in the CSF. This work marks an important step toward EPT validation, and highlights minimising CSF-specific errors as a priority for future work.
Philippa SHA (London, United Kingdom), Jierong LUO, Matthew CHERUKARA, Karin SHMUELI
13:30 - 15:00
#47955 - PG241 Fatty liver spectroscopy: is the analysis of more peaks the key to better diagnosis?
PG241 Fatty liver spectroscopy: is the analysis of more peaks the key to better diagnosis?
Metabolic associated fatty liver disease (MAFLD) is one of the most prevalent chronic liver diseases worldwide, closely linked to obesity, insulin resistance, and metabolic syndrome [1]. While liver biopsy is the gold standard, it is invasive, costly, and impractical for early detection. Using ex-vivo MRS and gaschromatography (GC-MS), previous studies have demonstrated that the composition of fatty acids in the liver changes as MAFLD progresses from simple steatosis to steatohepatitis. More recently, a study from our group, using MRS signal from the hepatic lipids and principal component analysis identified the 3 main groups (Healthy, steatosis steatohepatitis) by using as input the 7 peaks corresponding only to the fatty acids [2]. This approach shows promise for distinguishing between different stages of MAFLD based on metabolic shifts, providing a potential non-invasive diagnostic method. In this work we want to stablish : if we had access to more peaks (other metabolites/lipids,besides only fatty acids) the classification of MAFLD groups would improve?. Therefore the aim of this pilot study is to answer the question: Is the analysis of more peaks the key to better diagnosis?
The methodology involved analyzing lipids extracted from livers of eNOS KO mice fed a high-fat diet for 0, 4, 8, and 12 weeks (9 males and 10 females). Using a 9.4 T NMR scanner, 24 metabolic peaks—including 7 related to fatty acids—were identified and quantified. Data analysis focused on calculating the area under the curve (AUC) for each peak using NMR TopSpin software with predefined integral files for accuracy. Statistical comparisons between 7 and 24 peaks were performed using ANOVA in PRISM, analyzing data from 19 mice to identify key metabolic markers like olefinic and diallylic metabolites.
To interpret the data, Principal Component Analysis (PCA) was applied to reduce dimensionality and highlight key variations across disease stages, improving visualization. K-means clustering grouped samples based on metabolic profile similarities, allowing identification of distinct clusters corresponding to different NAFLD stages. The analysis showed significant decreases in olefinic and diallylic metabolites after 4 weeks of a high-fat diet(Fig.1), with levels stabilizing at 8 and 12 weeks. These metabolites exhibited an early decline followed by a plateau, highlighting their role as consistent metabolic indicators during disease progression. Other metabolites such as methyl, methylene, alpha, beta, and allylic showed no significant changes, limiting their usefulness for characterizing the disease.
PCA and K-means(Fig.2 and Fig.3) clustering revealed distinct groupings in metabolic profiles corresponding to the diet duration, supporting progressive stages of NAFLD. These analyses clearly separated samples by time points, with olefinic and diallylic metabolites as key contributors. The observed decrease in olefinic and diallylic metabolites is consistent with previous studies[3] reporting significant depletion of long-chain polyunsaturated fatty acids (LCPUFAs), particularly docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA), in NAFLD/MAFLD patients. These essential fatty acids, known for their multiple double bonds, are crucial components of cellular membranes and precursors for anti-inflammatory mediators. Their reduction likely underlies the diminished olefinic and diallylic signals detected in our MRS analysis, reflecting disrupted lipid metabolism and increased oxidative stress associated with disease progression.
The absence of significant variation in other metabolites highlights the value of targeting a refined subset of key metabolic markers to accurately characterize NAFLD. In this context, Principal Component Analysis (PCA) and clustering methods have proven effective for uncovering metabolic progression patterns and distinguishing disease stages by focusing on the most informative metabolites, thereby enhancing the specificity and interpretability of the analysis.
Focusing on a reduced set of 7 informative metabolic peaks improved clarity and reduced noise compared to analyzing all 24 peaks, which suffered from overlapping signals and redundancy. While the 24-peak model showed narrower intra-cluster variance—potentially beneficial in scenarios with higher experimental or biological noise—the 7-peak model achieved better separation of clinically relevant groups and yielded more interpretable results under controlled conditions. Therefore, we conclude that for this experiment, analyzing 7 peaks is sufficient, as analyzing 24 peaks does not provide additional meaningful information beyond what the 7 peaks reveal.
Catalina PARRA GARAFULIC (Ninguno, Chile), David TERRAZA, Laura MANJARES, Thomas EYKYN, Marcelo ANDIA, Aline XAVIER
13:30 - 15:00
#47744 - PG242 Understanding brain apparent T2 estimations with DESS sequences.
PG242 Understanding brain apparent T2 estimations with DESS sequences.
The Dual-Echo Steady-State (DESS) sequence can be used for T2 mapping [1]. This sequence involves low flip-angle radiofrequency pulses, making it an interesting alternative for imaging at high and ultra-high field due to Specific Absorption Rate limitations. The DESS sequence has recently been optimized for brain T2 mapping by mitigating artefacts due to breathing and underlying B0 field variations [2]. Despite the significant improvement and validation against a spin-echo reference in phantoms, the estimated apparent T2 remained biased in highly concentrated agar-based phantoms [2], suggesting magnetization transfer (MT) influences. Furthermore, in brain tissues, water compartmentation represents an additional intrinsic source of variabilities when considering a mono-compartment model to estimate T2. This study aims to combine numerical simulations and MRI acquisitions to understand the behaviors of T2 estimates from the DESS method.
Acquisitions were performed at 3 T (MAGNETOM Prisma, Siemens Healthineers, Erlangen, Germany) on one volunteer to estimate biophysical parameters required for the modeling and evaluate the consistency with numerical simulations.
The MRI protocol included a 3D DESS sequence for T2 mapping, complemented with an MP2RAGE for the required T1 prior. The T2 estimation with the optimized DESS protocols was performed as described in [2]. In addition, a quantitative MT (qMT) protocol was acquired following the methodology in [3] for the estimation of the Macromolecular Proton Fraction (MPF) from the binary spin-bath model, used as a surrogate of myelin water pool size (Myelin Water Fraction; MWF). A summary of the sequences’ parameters is provided in Table 1. DESS and qMT frameworks were both corrected for transmit field inhomogeneities.
T2 measurements were evaluated in representative regions of interest (ROI) from white (JHU probabilistic atlas [4]) and deep gray (recon-all from FreeSurfer [5]) matter, using the anatomical UNI image from the MP2RAGE acquisition.
Analyses were complemented by simulations to explain the experimental measurements. A realistic 4-pool model was used [6], considering two exchanging water reservoirs (namely myelin water [MW] and intra/extra-cellular water [IEW]), each of them exchanging with two bound reservoirs (myelin and non-myelin). Synthetic signals associated with the DESS sequence were generated using the Extended Phase Graph formalism [7], considering biophysical parameters from Manning et al. [6] (Fig. 1), and as a function of the MWF. Synthetic signals, taken as the summation of MW and IEW transverse magnetizations (as experimentally observed), were then fitted to the mono-component T2 model. As a first approximation, MWF and myelin/non-myelin bound pools were assumed of identical size (Fig. 1). To isolate the effect of exchanges and bound proton contribution to the signal build-up, simulations were also run by considering a pure water-only 2-pool model, while accounting or neglecting inter-pool exchanges. Experimental apparent T2 maps from the DESS configurations and MPF map are shown in Fig. 2, and averaged values from representative ROIs and associated simulation results in Fig. 3. Globally, the TR=30 ms configuration yielded higher apparent T2 values (spanning from 49 ms to 63 ms in WM, and 44 ms to 66 ms in GM) than the TR=15 ms one (42 ms to 53 ms in WM, and 42 ms to 59 ms in GM). Simulation results using the 4-pool model best captures the dynamic as well as variations with TR. Conversely, both water-only 2-pool models fail to predict the behaviors (opposite trend as a function of TR with ΔT2<0 ms for MPF>6.5%; Fig 3c), especially as myelination increases (predicted T2 values > 65 ms for MPF>7.5% overall). This preliminary work suggests that accounting for MT effects and water compartmentation using a 4-pool model may be essential to explain the apparent T2 estimations using a DESS sequence. In contrast, only accounting for water compartmentation fails to predict the associated trends with sequence parameters, emphasizing the importance of bound reservoirs on the DESS signal build-up.
Compensating for these confounding factors, however, would require extensive MT- & multi-compartiments-sensitized acquisitions to solve for a 4-pool model, which is currently unfeasible within clinically acceptable scan times, and calls for a consensus on the protocol design to mitigate variabilities. This study highlighted the impact of MT (and associated T1 relaxation), water compartmentation and exchanges in apparent T2 measurements estimated by DESS, drawing attention to the limitations of simplified biophysical models for the quantitative description of biological tissues. More volunteers are required so as to refine these results, and further experiments will be performed at 7 T to evaluate the impact of field-dependent variables such as T1 [8].
Emile KADALIE, Nadège CORBIN, Aurélien TROTIER, Olivier M. GIRARD, Émeline RIBOT, Lucas SOUSTELLE (Marseille)
13:30 - 15:00
#45684 - PG243 Comparison of Bi- and Tri-Component Approaches in the Analysis of Short-T2 Tissues: Application to the Achilles Tendon.
PG243 Comparison of Bi- and Tri-Component Approaches in the Analysis of Short-T2 Tissues: Application to the Achilles Tendon.
The purpose of this study was to evaluate different application scenarios of ultrashort echo time (UTE) magnetic resonance imaging for investigating short-T2 components in the Achilles tendon and to assess their impact on the characterization of the tendon’s compositional structure.
A custom time-interleaved 2D-UTE pulse sequence, as illustrated in Fig. 1, was implemented to enable the acquisition of up to 64 echo time images (1). Measurements were performed with and without fat saturation (FatSat). Ten healthy volunteers (age 33.3 ± 6.91 years) underwent UTE MRI of the Achilles tendon. Two different approaches were compared to address fat contamination: (A) a Bicomponent model with FatSat, and (B) a Tricomponent model using a fat separation technique. Additionally, the impact of magnitude versus complex data reconstruction on compositional assessment was evaluated.
Using multi-echo 2D-UTE images acquired at 64 echo times ranging from 60 µs to 32 ms, T2* relaxation decays were quantified with the following four data fitting models(2, 3):
a) Bicomponent magnitude data model
b) Bicomponent complex data model
c) Tricomponent magnitude data model
d) Tricomponent complex data model
All four models yielded amplitudes of the short (AS) and long (AL) T2* relaxation components. Based on the estimated parameters, the short (FS) and long (FL) component fractions, expressed as percentages, were calculated as (4): FS = 100xAS/(AS + AL) and FL = 100xAL/( AS + AL ) respectively.
Image acquisition was performed on a clinical whole-body 3T scanner (Magnetom Prisma, Siemens Healthineers, Erlangen, Germany). All experiments used a whole-body RF coil for transmission and a flexible four-channel phased-array coil for reception. Image reconstruction and data processing were conducted offline using custom routines implemented in MATLAB (The MathWorks, Natick, MA, USA).
All analyses were performed using R Statistical Software (version 4.4.1). Normality of data was assessed by Shapiro–Wilk test and Q-Q plots. A two-way repeated measures ANOVA was performed with probands as the within-subject identifier and data acquisition/reconstruction type as within-subject factors. Post hoc pairwise comparisons were conducted using estimated marginal means with Holm-adjusted p-values to control for multiple comparisons. In the statistical analyses, the following data acquisition/reconstruction type scenarios (i.e., post-hoc pairs) were considered:
a) FatSat/Magnitude vs. non-FatSat/Magnitude
b) FatSat/Complex vs. non-FatSat/Complex
c) FatSat/Magnitude vs. FatSat/Complex
d) non-FatSat/Magnitude vs. non-FatSat/Complex,
and P-values < 0.05 were considered statistically significant. Ten healthy volunteers (3 males, 7 females; mean age: 33.3 ± 6.9 years), with no history of Achilles tendon injury, participated in the study. Institutional approval was obtained, and all participants provided written informed consent prior to the study. Figure 2 shows representative images acquired with and without FatSat, along with data fitting results from all four models applied to a region of interest (ROI) selected in the mid-portion of the Achilles tendon (MID). The results from the ANOVA analyses are summarized in Tab. 1, and the short component ratios Fs calculated from different acquisition/reconstruction scenarios are presented in Fig. 3. Regardless of the Achilles tendon region, fat saturation, reconstruction type, and their interaction all had a statistically significant effect on the short component ratio (p < 0.05). The interaction between fat saturation and reconstruction type was not of primary interest and had a minimal effect—explaining only 0.6% of the variance in the INS region (p = 0.025), 4.7% in the MID region (p < 0.001), and 3% in the MTJ region (p < 0.001). In all tendon regions, fat saturation increased the short component ratio for both reconstruction types. Conversely, complex reconstruction consistently resulted in a higher short component proportion, with a more pronounced difference observed in the non-FatSat condition. The estimated marginal means and their standard errors were consistent with the differences in group means and their standard errors calculated from the original data. Both the data acquisition and reconstruction methods significantly impact the short T2* component ratio and should be considered when comparing results across studies.
.
Acknowledgements
This work was funded by Czech Science Foundation grant no. GF25-19984L. Also, this research was funded in whole or in part by the Austrian Science Fund (FWF) 10.55776/PIN5555423. We acknowledge the core facility MAFIL supported by MEYS CR (LM2023050 Czech-BioImaging), part of the Euro-BioImaging (www.eurobioimaging.eu) ALM and Multimodal Imaging Node (Brno, CZ).
Peter LATTA (Brno, Czech Republic), Veronika JANACOVA, Martin KOJAN, Lubomír VOJTÍŠEK, Aneta MALÁ, Zenon STARČUK, Vladimir JURAS
13:30 - 15:00
#47780 - PG244 Temperature Dependence of the Apparent Diffusion Coefficient in Aqueous Soy Lecithin Solutions at 3T.
PG244 Temperature Dependence of the Apparent Diffusion Coefficient in Aqueous Soy Lecithin Solutions at 3T.
Isotropic test fluids or gels with well-defined apparent diffusion coefficients (ADC values) are essential tools for testing and validating diffusion-weighted imaging techniques [1-5]. We have previously shown that aqueous soy lecithin solutions are a beneficial material for the construction of diffusion phantoms with tissue-like ADC values [6]. Soy lecithin added to water provides a wide range of adjustable ADC values without introducing spectral signals. In addition, soy lecithin is compatible with agarose, allowing simultaneous tuning of ADC and T2.
However, the behavior of aqueous soy lecithin solutions at different measurement temperatures has not yet been investigated. Temperature is a critical factor, as the molecular mobility of water – and therefore the ADC – is strongly temperature-dependent [5,7]. Consequently, measurements taken under different conditions may yield different ADC values.
To address this issue, we systematically investigated the temperature-dependent behavior of aqueous soy lecithin solutions. Solutions with varying soy lecithin concentrations (0-10% (w/v)) were prepared, and for each concentration, ADC values were measured at seven different temperatures ranging from 3°C to 37°C. Based on these results calibration curves were derived that can be used to correct temperature-related ADC variations.
Data acquisition and analysis:
MRI was performed on a clinical whole-body 3.0 Tesla scanner (MAGNETOM Prismafit, Siemens Healthcare, Erlangen, Germany) using an 18-channel body-array coil. Measurement temperatures were as follows: 3°C, 10°C, 15°C, 20°C, 25°C, 30°C, and 37°C. Image processing, including mapping and analysis, was performed offline in MATLAB (R2022b, MathWorks, Natick, MA, USA).
DW-MRI was performed using a readout-segmented echoplanar imaging sequence with 4 different b-values (0, 50, 500, 1000 s/mm2). TR and TE were set to 5000 ms and 51 ms, respectively. ADC maps were calculated from the acquisitions with multiple b-values using a log-linear fitting of the signal intensities.
Materials and sample preparation:
Soy lecithin (Carl Roth, Karlsruhe, Germany) was dissolved in deionized water by magnetic stirring for 10 minutes at 650 rpm. The concentrations of soy lecithin varied from 0-10% (w/v). After preparation, the solutions were filled into sterilized polypropylene tubes (Greiner Bio-One, Frickenhausen, Germany) and brought to the desired temperature in a heating cabinet. To keep the temperature stable during the measurement period, the solutions were placed in a thermally insulated polystyrene box for the measurements. The relationship between ADC and temperature was found to be mono-exponential (Eqs. 1) across all tested soy lecithin concentrations (Figure 1).
ADC [T]=a_C ∙ exp(b_T ∙ T) (1)
The growth rate b_T of the exponential fit was identical for all concentrations, indicating a uniform temperature behavior for all aqueous soy lecithin solutions (Table 1). In contrast, the intercept a_C varied with concentration, reflecting the concentration-dependent decrease in ADC. This relationship followed a biexponential trend, independent of the measurement temperature (Equation 2, Figure 2).
a_C = ADC [C_lec ]=A ∙ exp(-B ∙ C_lec) + C ∙ exp(-D ∙ C_lec) (2)
Based on these findings, a combined model was established to describe the ADC as a function of both soy lecithin concentration and temperature:
ADC [C_lec,T] = (A ∙ exp(-B ∙ C_lec) + C ∙ exp(-D ∙ C_lec)) ∙ exp(b_T ∙ T) (3)
This model accurately described the experimental data (R2 = 0.966) and reliably captured the effects of soy lecithin concentration and measurement temperature on the ADC value (Figure 3). In this work, calibration curves were derived to describe the temperature and concentration dependence of the ADC value in aqueous soy lecithin solutions. The resulting model provides a practical tool for correcting temperature-related ADC variations and may help to improve comparability of DWI measurements under varying experimental conditions.
Victor FRITZ (Tübingen, Germany), Siri RAUPACH, Fritz SCHICK
13:30 - 15:00
#46642 - PG245 Beyond the IVIM-MRI Biexponential model: Improved Detection of Concussion Effects via Triexponential IVIM-MRI.
PG245 Beyond the IVIM-MRI Biexponential model: Improved Detection of Concussion Effects via Triexponential IVIM-MRI.
Concussion, a mild form of traumatic brain injury (mTBI), is linked to subtle microstructural changes often missed by conventional imaging [1].
In this study, we applied intravoxel incoherent motion (IVIM) MRI to compare perfusion and diffusion between concussion patients and healthy controls (HCs), using both the standard bi-exponential model [2] and an extended tri-exponential model that adds a fast diffusion component, potentially reflecting free water or rapid microvascular flow [3].
The bi-exponential model estimates tissue diffusion (D), perfusion-related diffusion (D*), and perfusion fraction (F), while the tri-exponential model provides separate parameters for tissue (Ds, Fs), perfusion (Dp, Fp), and fast/free diffusion (Df, Ff).
We hypothesize that the tri-exponential model may offer greater sensitivity to post-concussion changes and aimed to identify IVIM-derived biomarkers distinguishing concussion from HC.
We recruited 20 HCs (9 F, mean age 29.6±6.5) and 20 individuals with mTBI (10 F, mean age 28.0±7.5). All participants completed the Montreal Cognitive Assessment (MoCA) [4]; the mTBI group also underwent Glasgow Outcome Scale-Extended (GOS-E) testing [5]. Diffusion MRI was acquired on a 3T Philips Ingenia using a modified IVIM protocol with 14 b-values (10–3000 s/mm²) and up to 12 directions per b-value. Preprocessing included denoising and motion, distortion, and eddy-current correction. Bi- and tri-exponential IVIM maps were generated using a custom Python script.
Test-retest reliability was assessed in 11 HC (4 F, mean age 31.8±6.6) using ICC [6] and Bland-Altman [7] analyses.
Voxel-based analysis was conducted in R, using a linear model adjusted for age, sex, and motion. Group-level statistics included pTFCE [8] and Benjamini-Yekutieli FDR correction [9]. Effect sizes were computed using Hedges' g [10], with α = 0.05 and power = 0.80 (large effect: g>0.99). Lower MoCA scores were found in the mTBI group (HC=28.5±1.7, mTBI=25.5±3.1; t=4.2, p<0.001), while the mean value of GOS-E for mTBI was 6.40±1.10, indicative of moderate to good recovery. No difference in age was observed (t=0.73, p=0.47).
Figure 1 (a) shows ICC distributions across IVIM metrics. Slow diffusion parameters, D (ICC = 0.72±0.22) and Ds (ICC = 0.77±0.18), had high reliability. In contrast, bi-exponential D* and tri-exponential Dp had poor reproducibility. Perfusion fractions showed high reliability for Fp (0.71±0.20) and Fs (0.69±0.21) and moderate reliability for F (ICC = 0.63±0.24) and Ff (0.55±0.23).
Figure 1 (b) presents Bland-Altman plots for best/worst BIAS subject (left and right panels, respectively). Metrics with high ICCs (D, Ds, and Fs) had narrow limits of agreement (LOA), indicating strong test-retest agreement. D* and Dp showed high variability and wide LOA.
Group comparisons, focusing on metrics demonstrating good reliability (ICC>0.50), revealed significant tri-exponential IVIM changes in the concussion group (Figure 2). Specifically, the slow diffusion coefficient (Ds) was reduced in concussed individuals compared to HCs, particularly within frontal and parietal regions. This reduction may indicate a loss of slow-diffusing components, potentially stemming from microstructural disorganization or diminished hindrance due to tissue swelling. Similar patterns of reduction were observed for the perfusion-related fraction (Fp) and the fast perfusion fraction (Ff). Conversely, the fraction associated with tissue diffusion (Fs) was elevated in the concussion group, suggesting changes in the tissue microenvironment that might enhance the relative contribution of restricted water diffusion, potentially linked to edema or microvascular changes.
Bi-exponential IVIM results (Figure 3) showed lower tissue diffusion (D) in concussion subjects relative to controls in parietal/insula/temporal regions; minor D increments were observed but lacked substantial effect sizes. The perfusion fraction (F) was significantly lower in the concussion group, suggesting reduced microvascular perfusion post-concussion. A comprehensive summary of all findings is presented in Figure 4. Our findings support the utility of IVIM MRI, particularly the triexponential model, in detecting subtle brain changes after concussion. The triexponential metrics showed higher test-retest reliability and revealed more widespread group differences through voxel-based analysis.
Reduced Ds in concussion may reflect microstructural disruption or decreased restricted diffusion, while elevated Fs suggests changes in the microvascular or interstitial environment. Additionally, decreased Fp and Ff can be connected to tissue damage, edema, and cell loss. The biexponential results also showed lower F metric in concussion, indicating possible vascular alterations. Overall, these IVIM-derived metrics, especially from the triexponential model, may serve as promising biomarkers for concussion-related brain changes.
Maurizio BERGAMINO (Phoenix, USA), Lauren R. OTT, Molly MCELVOGUE, Ruchira JHA, Cindy MORENO, Radia WONG, Ashley M. STOKES
13:30 - 15:00
#45955 - PG246 Gadolinium Retention Dynamics in Polysaccharide Matrices: Exploring the masking effect for Gd ions in polysaccharide-rich environments.
PG246 Gadolinium Retention Dynamics in Polysaccharide Matrices: Exploring the masking effect for Gd ions in polysaccharide-rich environments.
The long-term fate of gadolinium (Gd) from GBCAs in the human body remains a subject of scientific investigation. While these agents have revolutionised diagnostic imaging, the potential for Gd retention in tissues raised concerns worldwide several years ago through unexpected hyperintense signal areas in clinical scans. This study investigates the interactions of Gd with biological molecules, focusing on the role of sugar structures in Gd retention. Using dextran sulphate (DS) as a model compound, we aim to elucidate how different molecular weights of polysaccharides influence Gd behaviour and detectability. Our investigation provides insights into the chelation process between Gd ions and sugar molecules, molecular weight dependent affects Gd relaxivity and potential mechanisms of Gd shielding that may affect MRI detection. This research contributes to the growing field of knowledge on Gd deposition and aims to improve our understanding of the safety of MRI contrast agents. By exploring these fundamental interactions, we are paving the way for more accurate assessment of Gd retention and potentially improved contrast agent designs in the future.
All MR measurements were performed on a 9.4 T MRI system (Bruker, Germany). T1 measurements were performed using a dephasing recovery sequence consisting of 50 π/2 pulses with subsequent gradient spoiling and image acquisition. R1 values were calculated from ROI-averaged values from R1 maps. To model the transchelation processes of Gd-ions to different sugars, DS (8 kDa – 500 kDa) in combination with GdCl3 were used as a model system. Light scattering measurements were used to investigate the increase of the aggregate size as a result of Gd-ion binding to DS and CSA. An increase in R1 was observed after the binding of Gd-ions to the added polysaccharides. Subsequently, a reduction of R1 could be observed with increasing polysaccharide concentration. Figure 1 shows R1 as a function of the concentration ratio between DS with different MW (with adjusted to total number of sugar units) and Gd-ions in solution. In all measurements, R1 increases from 0.6 s-1 (R1 of 25 μM Gd3+ in H2O) to ~0.8 s-1 and to ~0.85 s-1 for (DS) of MW = 8 kDa, 20 kDa and 500 kDa, respectively. The maxima are reached at ratios of ~101.1, ~101.3, and ~102.3. Subsequently, R1 decreases to ca. 0.4 s-1, 0.45 s-1, and 0.5 s-1 at ratios of ~103.1, ~104, and ~105 for (DS) of MW = 5, 20, and 500 kDa, respectively. Glycosaminoglycans (GAGs) display a heterogeneous distribution within tissues, thereby creating local microenvironments with the potential to act as hotspots for Gd deposition. The results obtained at first support the hypothesis that Gd binding to sugar structures, increases longitudinal relaxation rates (R1) thus providing a plausible explanation for clinical observations of tissue hyperintensities. This phenomenon can be attributed to the lower tumbling rate of Gd-sugar complexes in comparison to free Gd ions, resulting in enhanced relaxivity at low [GAG]:[GdCl3] ratios.
However, at higher sugar concentrations, this initial increase in R1 is counteracted by a loss of signal. This shielding effect is attributed to sulphate groups occupying Gd coordination sites, thereby limiting water access to the paramagnetic center. At the same time, Gd-mediated cross-linking of the sugar chains, as shown by light scattering data, induced aggregation and amplifies particle size. It is noteworthy that DS structures with lower molecular weight (MW) accelerate this masking phenomenon, presumably due to steric constraints that promote tighter coordination and faster saturation of binding sites.
This MW-dependent shielding may lead to a potential underestimation of uncleared Gd3+ in clinical MRI, as aggregated complexes may avoid detection despite persistent tissue deposition. These findings underscore the necessity to refine imaging protocols and analytical methods to account for polysaccharide-Gd interactions, particularly in tissues with high GAG concentrations. It is recommended that future studies investigate how variations in GAG composition and sulphation patterns affect Gd retention and visibility, thereby addressing a critical gap in understanding long-term deposition. Our results show that the binding of Gd ions to polysaccharides and the associated increase or decrease in R1 is strongly dependent on the molecular weight (MW) in addition to the concentration ratios. Using model structures such as DS, we have shown that in relaxometry experiments, in addition to hyperintense signals, a clear masking of the Gd ions can be caused by the polysaccharide concentration. We were able to demonstrate this effect for different MWs and showed that the signal gain and loss is characteristic for each individual molecular size. This highlights the need for further research in this area to ensure that the problem of deposited Gd ions in biological tissues is not underestimated in the future.
Patrick WERNER (Heidelberg, Germany), Matthias TAUPITZ, Leif SCHRÖDER
13:30 - 15:00
#47681 - PG247 DTI-EPI sequence optimization on a preclinical nanoScan 7T MRI scanner using an anisotropic diffusion phantom.
PG247 DTI-EPI sequence optimization on a preclinical nanoScan 7T MRI scanner using an anisotropic diffusion phantom.
Phantoms, as reference tools for quantitative MRI, maintain data reproducibility by ensuring scanners correct operation and by leading to improvements in image quality. They allow development and validation of new methods and comparison between acquisition and analysis performances[1–3]. Diffusion phantoms are more commonly developed consider isotropic diffusion[4–9]. Recent literature shows only a few examples of anisotropic diffusion phantoms in more sophisticated and updated versions[10–12], considering in few cases also different fiber crossing angles or a different fiber density rate[13]. Furthermore, few recommendations are available for preclinical purposes[2] and thus less studies focus on diffusion sequence optimization, giving a reference for many possible ex-vivo/in-vivo conditions. The aim of this preliminary study is to optimize a DTI-EPI sequence, exploiting a preclinical diffusion phantom, considering DTI metrics, image quality in terms of SNR maximization and tractography quality as target of optimization.
An MRI diffusion phantom produced in a preclinical custom-made size was used (Phantom Metrics, Psychology Software Tools, Inc., Pittsburgh, PA, USA). The phantom consisted of a 4mm x 4mm x 20mm nylon fiber bundle (Taxon™ technology[14]). Phantom manufacture: 1,470 round fibers (diameter range = 105–110μm, 6,532 water filled Taxon holes (inside diameters = 0.68-0.78μm) per fiber, hindered water spaces between the round fibers estimated as 8% of the fiber bundle total volume. The phantom was placed in a water filled 50ml falcon tube, near the tube surface. Diffusion imaging was performed on a Mediso nanoScan®7T MRI scanner with a DTI-EPI sequence. Sequence parameters were tuned on the phantom, keeping a resolution of about 200μm, 25 diffusion directions and b=500s/mm2. Main changes were focused on single/multi-shot choice, TE/diffusion time trade off, imaging and diffusion gradient parameters as well as encoding direction to minimize artifacts and number of excitations. Image Signal to Noise ratio (SNR) maximization has been considered first target of sequence optimization. Data processing was performed through the ExploreDTI tool[15]. After data reconstruction from raw diffusion-weighted images and LPCA Denoising Algorithm application[16], ADC, FA, MD and AD maps were calculated. The known fiber density and direction and thus the diffusion behavior have been taken as reference to select the best parameters setting, while further considerations have been driven about image and tractography quality. The first step of parameters optimization resulted in the following set: amplifier configuration = 750V, 4 shots, 80% symmetric Partial Fourier (PF) acquisition (0.22x0.28mm2) in the phase encoding direction, diffusion gradient ramp time = 1.016ms, duration(δ)=1.2ms, and interval(Δ)=9ms. With respect to other considered parameter settings, this configuration became the reference since it gave the best results in terms of FA (0.72), as precise representation of the phantom anisotropic nature (tractographic reconstruction shown in Fig.1). DTI metrics obtained in function of parameters variation are reported in Table 1. Starting from the reference setting, image and tractography quality have been quantified using number of reconstructed tracts (Fig.2) and SNR calculation (Fig.3) respectively. Both indices have been considered in function of number of excitations (NEX 2 to 5), number of shots (1,2,4), two PF types and when applying or not the denoising algorithm. The reference setting resulted to be the best parameters configuration, with metrics comparable to literature values[17]. The higher FA together with the lower AD confirm a high diffusion along the known direction of the fibers, more than in the axial one. Using a different amplifier configuration and adjusting the diffusion gradient parameters, high FA is obtained when ramp time and duration are similar and when applying the symmetric 80%PF in case of 4 shot and asymmetric in case of 2 shots. A sensible FA reduction can be noticed especially when using a 1 shot. Indeed, even if the acquisition time decreases when using 1 shot and could be better in terms of in-vivo applications, the echo time considerably increases, producing less signal from the phantom and therefore low SNR. Increasing NEX confirmed an increase of SNR values, with a more evident trend when considering the non-denoised images, even if for these latter, absolute values 10 times lower than the corresponding ones in denoised images can be noted. The same clear trend appears also in tractography quality for the non-denoised images, while a better reconstruction in terms can be achieved even for the 1 shot but applying the denoising algorithm. No general substantial differences are produced between the 2 PF types. In this preliminary study, a reference optimized sequence has been obtained, with improvements in term of DTI metrics, after acquisitions with different parameters sets.
Valeria CERINA (Milan, Italy), Jozsef SINKO, Magor BABOS, Alfonso MASTROPIETRO, Rosa Maria MORESCO, Gergo BAGAMERY
13:30 - 15:00
#47841 - PG248 Contrast Adjustment for MR Images at 7 T.
PG248 Contrast Adjustment for MR Images at 7 T.
MRI is capable of generating a variety of contrast mechanisms. The acquired signal used to generate the image depends on a range of parameters, where each of them is either object specific or method specific. Knowledge of this dependency provides the possibility to adjust the signal intensity of any spin isochromat by modifying method specific parameters in the dependency. As this reliance is known for a sequence that is employed to generate the image, the signal intensity of and therefore the contrast between different isochromats in MR images are adjustable. In this work we present proof of this concept to UHF-MRI (7 T) aiming for applying it to knee imaging.
The signal generated by Spin-Echo (SE) sequence [1] was simulated using in-house MATLAB [2] scripts based on closed-form solution of the Bloch equations for SE signal. Considering a specific range of TRs and TEs given to the simulation while other parameters are known, the outcome is the range of TRs and TEs at which a certain level of contrast between two spin isochromats will be generated.
Attaining the desired level of contrast was verified on a phantom. The phantom consisted of an apple, a potato and a citrus. Images were acquired using SIEMENS 7T MR Imager (MAGNETOM Terra) and dedicated knee coil (1Tx 28Rx Knee Coil 7T Clinic). Due to images of each of the three objects being not isointense, relatively homogeneous regions were selected for relaxation times calculation. Using inversion recovery acquisition method (TIs=40, 100, 300, 600, 1000, 1500, 3000, 5000 ms, TRs=TI+10000 ms) and an exponential fitting to the acquired data, we estimated the T1 value of apple and potato in a relatively homogeneous region (7 slices (apple), 19 slices (potato), 30 slices (total),2.5/0.5 mm thickness/gap). SE acquisition method applied at multiple TEs (TEs=6, 8, 12, 15, 30, 45, 65, 95, 120, 150, 180 ms, TR=7500 ms) and an exponential fitting were performed to measure T2 of the two objects in almost the same regions (8 slices (apple),18 slices (potato), 40 slices (total), 2.5/0.5 mm thickness/gap). SE acquisition with very short TE and very long TR (TE=6 ms, TR=15000 ms ) to leave the signal weighted only by proton density was used where relative signal intensities of the two objects from regions of equal size delivered relative spin density which was then averaged across selected slices. The contrast between apple and potato was calculated in two slices (2.0/0.1mm thickness/gap) which were adjacent and among the slices where relaxation times were calculated.
Our evaluation of contrast adjustment was performed in two sets of SE imaging, one with long TR=5580 ms (with five contrast levels from 95±5% down to 15±5%) and the other with relatively shorter TR =1780 ms (with four contrast levels from 95±5% down to 65±5%). By feeding the simulation with the relaxation times, spin density ratios of the two objects, and the TR and the flip angle of the sequence refocusing pulse, we calculated TEs at which the contrast lies within the desired level and range. The range of TEs given to the simulation was 10 to 120 ms with resolution of 50 points equally spaced. These TEs were given to the sequence to generate images with adjusted contrast. Figure 1 shows the result of simulation for a specific TR of 5580 ms. For each level and range of contrast the calculated TEs are presented in the corresponding plot. The measured relaxation times of the two objects and their relative proton density ratios normalized to that of apple are given in Table 1. Tables 2 and 3 manifest the desired levels of contrast, the TEs that were calculated and entered into the SE sequence for each level, the measured CNR and its level relative to the reference (ref) for two sets of SE image acquisitions with different TRs. The levels are given in percentage relative to the maximum achievable contrast. Measured mean CNRs lie within the range of 10% which was set for each level of contrast. This work is an initial step towards generating contrast between tissues at any desired level in MR images, e.g. in knee MR images generated at UHF (Ultrahigh Field). To achieve this, all parameters that affect the MR signal intensity must be considered in the whole process of contrast adjustment. The type and number of some of these parameters are dependent on the sequence which is employed to generate the images. The accuracy of the calculations could also be affected by the applied reconstruction technique e.g. when parallel imaging is employed to accelerate the acquisition. Knowledge of precise values of relaxation times of the spin isochromats that are imaged elevates the accuracy of the contrast adjustment. In this work we present a proof of concept of the possibility to gain specific levels of contrast between different objects in MR images with the support of simulation. The approach could be applied to a variety of sequences and paves the way for establishment of a fully automated contrast adjustment for MR images.
Seyedmorteza ROHANI RANKOUHI, Mikhail ZUBKOV, Laurent LAMALLE, Mohamed Ali BAHRI, Christophe PHILLIPS (Liège , Belgium)
13:30 - 15:00
#47674 - PG249 Assessment of the CEST SSFP-FID sequence performance on the nanoScan 7T MRI scanner.
PG249 Assessment of the CEST SSFP-FID sequence performance on the nanoScan 7T MRI scanner.
Chemical Exchange Saturation Transfer (CEST) MRI is a promising molecular imaging tool based on proton exchange between the target molecule and water. Because of its unique contrast mechanism, the interest for this technique is still increasing for both clinical and preclinical applications. Many molecules can act as CEST agents, both endogenous molecules and exogenous agents. APT (Amide Proton Transfer) CEST is one of the most widely used CEST techniques. APT signals primarily arise from the proteins and peptides found in tissue[1]. The technique is most commonly used for detecting cancer[2, 3] and ischemic stroke[4]. CEST MRI of iopamidol offers a noninvasive method for assessing tissue pH by leveraging the pH-sensitive properties of iopamidol contrast agent[5–7]. This approach provides valuable diagnostic insights, especially for detecting pathological conditions such as tumors[8]. Our aim was to investigate the performance of CEST-SSFP-FID[9] sequence in terms of APT and iopamidol CEST using phantoms of poly-L-lysine (0.1% w/v) and iopamidol (30mM), respectively.
All measurements were performed on a nanoScan® 7T MRI Mediso scanner. For each CEST-SSFP-FID acquisition, the following parameters were used: TE=1.85ms, TR=3.71ms, matrix = 64 × 64, FOV = 38.4 mm x 38.4 mm, slice thickness = 2mm (single-slice), flip angle=30°, NEX=1. Centric k-space filling was applied with 10 dummy scans. Presaturation delay of 5s was set between the readout and the following CEST saturation pulse. The total scan time was 2min 37s to acquire CEST images at 28 saturation frequencies. Three CEST saturation conditions have been investigated. The use of different number of pulses (single or train of 10 hard pulses with interpulse delay of 10µs) during the same total saturation time. Then, the effect of saturation power (B1). Lastly, the application of a spoiler gradient after the saturation. We acquired 28 saturation frequencies for both substances, with iopamidol ranging from -8ppm to +8ppm and poly-L-lysine ranging from -6ppm to +6ppm. For both phantoms, less points have been acquired in the negative range, since the target molecules have no influence on water signal in that range. A reference image was obtained with saturation frequency at -150ppm, where no CEST effect was expected. Z-spectra were obtained from a circular ROI placed in the center of the phantom and avoiding artifacts at air-phantom border. Image intensities were averaged within this ROI and expressed as a percentage of the ROI-averaged values obtained from the reference image. These values have been used to generate the Z-spectra and to calculate the asymmetric magnetization transfer ratio (MTRasym)[10] for iopamidol at 4.2ppm and 5.6ppm and for poly-L-lysine between 2 and 4 ppm. Figures 1 and 2 show Z-spectra obtained with different saturation powers for iopamidol and poly-L-lysine, respectively. Higher contrast peaks are expected at 4.2ppm and 5.6ppm for iopamidol and at an extended area between 2 and 4 ppm for poly-L-lysine. Figure 3a and 3b show Z-spectra obtained using 3µT saturation power for both iopamidol and poly-L-lysine. Figure 3a shows the effect of switching on and off the spoiler gradient after CEST saturation pulse, obtaining no evident difference in the Z-spectra and MTRasym values for both substances. Figure 3b focused on the effect of pulse train compared to single pulse. As expected, higher loss of signal on Z-spectra as well as higher MTRasym values (as shown in Figure 4) correspond to power value increase. However, the peak width also increases and this could make the target effect less detectable. For CEST contrast of iopamidol, 3µT can be a good trade-off, since it produces high contrast at 4.2ppm and 5.6ppm, keeping the water peak sufficiently narrow. Additionally, the OH- group appears more evident with 3µT compared to 6µT. For CEST contrast of poly-L-lysine, 3uT is also a good trade-off.
No substantial difference can be noticed in Z-spectra and MTRasym values for both substances, demonstrating the high quality of the pulse train produced by this instrument. Typically, the maximum contrast for APT CEST happens at 3.5ppm, but acquiring poly-L-lysine on this scanner the signal loss in Z-spectra and MTRasym values result higher at 2.5ppm for each saturation conditions. CEST-SSFP-FID sequence is a fast and reliable method to measure the CEST effect of APT or iopamidol. The CEST application package provided by Mediso on the nanoScan® 7T scanner was validated on phantom scans and the CEST saturation pulse could be optimized by using poly-L-lysine and iopamidol, so in-vivo studies can be carried out in the future.
Valeria CERINA (Milan, Italy), Virag DARANYI, Magor BABOS, Gergo BAGAMERY, Mark David PAGEL
13:30 - 15:00
#45871 - PG250 Enabling preclinical, quantitative, in-vitro studies of hypoxia with MRI with novel application of microfluidic chips.
PG250 Enabling preclinical, quantitative, in-vitro studies of hypoxia with MRI with novel application of microfluidic chips.
Hypoxia is a cancer biomarker indicating tumor aggressiveness. Incentive exists to use non-invasive MRI technology to map and quantitatively measure hypoxia. However, traditional in-vitro hypoxic models are incompatible with the MRI experiments that are needed to develop MRI-based hypoxia quantification. Microfluidics technology creates size-controlled 3D cell spheroids wherein the level of hypoxic condition varies with the spheroid size. Using hypoxic spheroids for MRI-based determination of hypoxia has not been investigated. The objective of this research was to demonstrate the viability of using hypoxic cancer spheroids in microfluidic chips to quantify hypoxia using MRI.
Microfluidic chips were utilized to seed, form and culture human colon cancer (HCT116) cells using techniques previously established (Refet-Mollof et al, 2021). A total of 120 hypoxic spheroids (~ 700 µm diameter) and 120 normoxic spheroids (~ 250 µm diameter) were formed; the size of the spheroids was verified with microscopy. The proportion of silicon to media or cells in the chips was constant. A western blot (the gold standard of hypoxia quantification) was performed to quantify the hypoxia-inducible carbonic anhydrase IX (CAIX) protein in half of the normoxic and half of the hypoxic spheroids, verifying the hypoxic content prior to the experiment, and also demonstrating the level of hypoxia in the hypoxic versus normoxic cells. The chips with remaining spheroids were scanned in a 3 Tesla Siemens MRI machine. AT2-weighted sequence (resolution of 0.8 x 0.8 x 0.7 mm3) was employed. Medical Image Merge (MIM, version 7.2.8) was used to measure the signal from each spheroid in the microfluidic chip from a 1 mm diameter region-of-interest. The signals from both normoxic and hypoxic spheroids were separately averaged; standard deviations were calculated. A Student’s t-test was applied to assess significant differences in signals between the normoxic and hypoxic spheroids. After the scans, the CAIX levels in the scanned chips were measured with western blot to verify that hypoxia was maintained throughout the MRI scan. Individual wells (~ 1 mm) were resolved on the MRI images, indicating the feasibility of using microfluidic chips for cellular studies with MRI. Figure 1 displays an MRI image of the hypoxic spheroids in the wells, illustrating their resolution on MRI. The hypoxic spheroids yielded significantly less signal compared to the normoxic spheroids (normoxic: 1530 +/- 294, hypoxic: 1425 +/- 269, p < 0.05). The loss in signal is expected given that the hypoxic condition decreases T2 relaxation times. Western Blot results confirmed the hypoxic condition during the MRI scans and that the hypoxic spheroids had more CAIX protein than the normoxic spheroids. This research demonstrates the feasibility of using microfluidic chips for in-vitro studies of hypoxic cells with MRI, as well as other cellular studies. Further studies will explore using additional MRI sequences, quantifying various levels of hypoxia, and developing complex materials using microfluidic chip technology. Eventually, the research will be expanded to cellular models that more closely resemble human anatomical systems, while maintaining the ability to quantitatively study hypoxia. A T2-weighted MRI sequence yielded significantly less signal from hypoxic spheroids versus normoxic spheroids in microfluidic chip wells, unlocking the potential for using microfluidic chips in cellular studies of hypoxia and other conditions.
Clara J FALLONE (Calgary, Canada), Ali GOLESTANI, Yang YANG, Elena REFET-MOLLOF, Rodin CHERMAT, Thomas GERVAIS, Philip WONG, Leila LARIJANI, Karl RIABOWOL
13:30 - 15:00
#47737 - PG251 MRI-Based 3D Modeling of the SOD1 Mouse.
PG251 MRI-Based 3D Modeling of the SOD1 Mouse.
The ethical and sustainable optimisation of animal preclinical studies has becoming an increasingly important matter for enhacing the planning and translational validity of in vivo experiments. The development of in silico realistic anatomic plays a crucial role in addressing this issue. In this study, we present a high-resolution, full-body 3D mouse model designed from magnetic resonance imaging (MRI) scans, to support simulations of non-invasive trans-spinal direct current stimulation (tsDCS) in mice.
Acquisition of MRI data: MRI data were acquired from an ex-vivo male C57BL/6 mouse (sacrificed under the Regierungspraesidium Tübingen animal experimentation license no 1522) at postnatal day 30 (PND 30), using an ultra-high-field 11.7 Tesla small animal MRI system (BioSpec 117/16, Bruker BioSpin, Ettlingen, Germany). To visualize tissue interfaces and structures with susceptibility contrast, FLASH T2* acquisition protocol was applied: TR/TE = 1800/4.25 ms, flip angle = 25.0°, matrix size = 250 × 230, spatial resolution = 100 × 100 × 350 µm³, bandwidth = 65.8 kHz, and 150 signal averages, yielding a total imaging duration of approximately 17 hours and 15 minutes. A total of 165 axial slices were acquired. An example of a resulting MRI axial slice is presented on Figure 1. This contrast allowed a visible distinction of key anatomical structures of our model, specifically the following spinal regions: grey matter (GM), white matter (WM), cerebrospinal fluid (CSF), dura mater, intervertebral discs, and vertebrae.
Design of the in vivo mouse model: Segmentation of relevant anatomical structures (skin, spinal GM and WM, intervertebral discs, vertebrae, skull and brain, and internal organs) was performed semi-automatically using ITK-SNAP (www.itksnap.org) threshold-based method [1]. For the spinal GM and WM, vertebrae, and visceral organs, manual regions of interest (ROIs) were defined at each relevant region along the cervical, thoracic, and lumbar segments of the spinal cord. The segmented data were converted into surface meshes (.STL file format). Tissue masks were converted into 3D surface meshes and imported into Blender 4.0 (www.blender.org/) and 3-MATIC module from MIMICS software (v16) (www.materialise.com/en/industrial/software/3-matic), for manual refinement, including smoothing, remeshing, and boolean operations to prevent intersections. Additional anatomical structures, such as CSF, dura mater, subcutaneous fat, and muscle, were modelled using defined offsets based on MRI measurements and literature values [2]. All final surface meshes are shown in Figure 2. The resulting model offers full-body anatomical coverage with a focus on the spinal region, allowing for detailed spatial characterisation of electric field (EF) and direct current (DC) distribution under multiple tsDCS electrode configurations. This virtual platform enables the simulation of stimulation parameters and biological tissue electric properties, facilitating parameter optimisation prior to in vivo experimentation, ensuring both the safety and efficacy of tsDCS protocols [3–6]. To our knowledge, this is the first high-resolution, full-body 3D model of a small animal to be applied in the context of non-invasive spinal cord stimulation. Previous studies have predominantly focused either on larger animal models, such as the rat or cat [5,6], or have been limited to invasive stimulation techniques [7–9]. The need for a full-body anatomical model is particularly relevant, as it allows simulation of multiple electrode montages and current pathways, accounting for other anatomical features that influence EF distribution on the spinal cord. This is essential for accurate prediction and optimisation of tsDCS protocols.
Our model was integrated in the first in silico-in vivo study to predict spatial distribution and geometric constraints of the EF resulting induced by tsDCS in SOD1 mice, the established animal model of Amyotrophic Lateral Sclerosis, anatomically similar to the C57BL/6 mice. This work provides a foundational tool for the translational modelling of neuromodulation in small animal models, bridging preclinical and clinical research. This computational approach contributes significantly to the 3Rs (Replacement, Reduction, and Refinement) in animal research. Future developments further validation with in vivo electrophysiological data, and full pipeline automatization, spanning MRI segmentation, 3D anatomical modelling, and FEM-based simulation, enabling reproducible and scalable generation of subject-specific models.
Leonor DE OLIVEIRA PIRES (Lisbon, Portugal), Alireza ABAEI, Francesco ROSELLI, Marcin BĄCZYK, Sofia Rita FERNANDES
13:30 - 15:00
#47912 - PG252 Deep Learning-Based Denoising of Preclinical MRI: A Preliminary Study of Autoencoders on T2-Weighted Images.
PG252 Deep Learning-Based Denoising of Preclinical MRI: A Preliminary Study of Autoencoders on T2-Weighted Images.
Magnetic resonance imaging (MRI) plays a central role in preclinical neuroscience by enabling non-invasive visualization of structural and functional changes in animal models [1–2]. High-resolution images with elevated signal-to-noise ratio (SNR) allow precise anatomical assessments and support longitudinal studies [3]. However, challenges such as motion artifacts, field inhomogeneities, and prolonged acquisition under anesthesia persist—even at ultra-high field strengths like 7 T [1,4]. Image noise, in particular, can obscure subtle anatomical features and compromise measurement accuracy, making effective denoising essential for reliable analysis.
Traditional methods, including Gaussian filtering and median smoothing, often suppress noise at the expense of structural detail. More advanced techniques like Non-Local Means (NLM) and BM3D improve performance by exploiting image redundancy but are computationally intensive and sensitive to parameter settings [5,6].
Recent advances in deep learning have demonstrated superior denoising capabilities by learning noise patterns directly from data [6]. Convolutional autoencoders (CAEs) reduce noise by encoding images into latent representations that preserve key anatomical structures and reconstruct clean images using pixel-wise loss optimization [7].
In this study, we evaluate the performance of the autoencoder architecture in denoising T2-weighted MR images of rat brains, aiming to enhance image quality while maintaining anatomical accuracy.
16 female Wistar rats were imaged using a 7T preclinical MRI system (MR Solutions Ltd., Guildford, UK). Animals were anesthetized with 1–2% isoflurane and immobilized using a stereotaxic head holder with ear and bite bars to minimize motion. Physiological monitoring included respiration and body temperature (Model 1030, SA Instruments, NY, USA). The imaging protocol included T1-weighted (FSE; TR = 1000 ms, TE = 11 ms, resolution: 0.13 × 0.13 × 0.8 mm³) and T2-weighted (FSE; TR = 2500 ms, TE = 40 ms, resolution: 0.14 × 0.27 × 1 mm³) acquisitions.
Clean images were treated as ground truth. Artificial noise—Gaussian, Rician, Rayleigh—was added to generate paired noisy datasets. The dataset was randomly split into training (70%), validation (15%), and test (15%) sets. Data augmentation was applied, including random flipping and rotation. All images were pre-processed through filtering, resizing, and normalization to ensure consistency.
A convolutional autoencoder deep learning model was implemented. The model was trained using paired noisy-clean data. Model performance was evaluated using loss function, peak-signal-to-noise-ratio (PSNR), structural similarity index (SSIM), contrast-to-noise-ratio (CNR), and mean squared error (MSE). In this preliminary study, a convolutional autoencoder was trained to denoise artificially corrupted T2-weighted brain MR images from a dataset of 16 rats. The noisy input images had an average PSNR of 22.40 dB, SSIM of 0.4653, MSE of 0.006571, and CNR of 2.7138.
Following 50 epochs of training, the model was evaluated on a separate test set, achieving an average PSNR of 28.50 dB, SSIM of 0.7273, MSE of 0.001435, and CNR of 3.5887. The evolution of validation performance during training is summarized in Table 1. Consistent improvements in PSNR, SSIM, and CNR, along with a decrease in MSE, were observed throughout the learning process.
Figure 1 presents the training and validation loss curves, while Figure 2 shows the progression of PSNR values over epochs. This study demonstrates the feasibility of using convolutional autoencoders (CAEs) for denoising T2-weighted rat brain MRI. The model yielded consistent improvements in PSNR, SSIM, and CNR, indicating effective noise reduction with preserved structural detail. These findings align with recent work highlighting the advantages of deep learning-based denoising over traditional filtering methods in preclinical neuroimaging [4,8].
One limitation of this study is the use of JPEG-formatted images, which may introduce compression artifacts and loss of structural fidelity. Additionally, the model was trained on synthetically noised images without a true ground truth. To address this, future studies should consider phantom-based validation to generate reference data with known signal and noise properties, enabling more accurate and reproducible assessment of denoising performance.Further research is needed to validate performance on raw scanner data and to compare CAEs with alternative deep learning architectures such as GANs. This study shows that convolutional autoencoders (CAEs) can effectively improve image quality in T2-weighted small animal brain MRI under synthetically induced noise. The model achieved notable gains in PSNR, SSIM, and CNR, demonstrating its capacity to suppress noise.
Fatma Beyza KULA YEŞILOT (İstanbul, Turkey), Leen HAKKI, Mehmed ÖZKAN, Oğuzhan HÜRAYDIN, Uluç PAMUK, Esin ÖZTÜRK-IŞIK, Pınar Senay ÖZBAY
13:30 - 15:00
#46408 - PG253 Quantifying the Impact of Formalin-Fixation on R2* Orientation Dependency in Human Brain White Matter.
PG253 Quantifying the Impact of Formalin-Fixation on R2* Orientation Dependency in Human Brain White Matter.
The orientation of nerve fibers relative to the main magnetic field (B0) of the MRI system has been shown to significantly affect the apparent transverse relaxation rate R2* [1–4]. Many existing studies thereby rely on formalin-fixed brain tissue [5,6]. However, fixation is known to substantially alter relaxation parameters [7,8]. In this study, we investigated the effect of formalin fixation on the orientation dependency of R2*.
MRI brain scans were performed on a Siemens MAGNETOM Prisma 3T system with a 20-channel head and neck coil (Siemens Healthineers, Erlangen Germany). The dataset comprised 22 individuals and was categorized into three acquisition groups: in-vivo, postmortem (PM) in-situ (brain not excised), and PM formalin-fixed ex-situ (brain excised).
The in-vivo group consisted of four healthy volunteers (age 29.5 ± 5.6 y; 4 f). The PM in situ group included eleven neurologically healthy deceased individuals (age at time of death 59.4 ± 15.2 y; 4 f, 8 m), as well as six deceased patients with ALS (age 63.2 ± 8.2 y; 6 m). The brains of the ALS patients were excised the day after the in-situ scan and fixed in formalin containing 4.5% formaldehyde (acid-free, phosphate-buffered, Roti-Histofix 4.5%, Carl Roth, Karlsruhe, Germany) at room temperature for 3 months for a subsequent PM ex-situ MRI scan. For the PM ex-situ MRI examination, the excised brains were placed in a spherical acrylic container. To prevent deformation and movement of the brain, custom-made brain-specific plates were 3D printed [7]. After placement of the brains in the containers, they were filled with a proton-free and tissue susceptibility-matched fluid (Galden SV 80, Solvay Specialty Polymers, Tavaux, France) to improve ex-situ MRI scanning conditions [9]. MRI sequences included MP2RAGE or TSE (brain tissue segmentation) [10], DTI with b = 2000 s/mm2, 3 b = 0 s/mm2, N = 64 isotropic (calculation of nerve fiber angle) and multi-echo GRE with 12 TEs, TE1= 5.79 ms, TR = 68 ms (R2* calculation).
Data analysis was performed using the FMRIB Software Library (FSL v6.0.7.13) [11–13]. Skull stripping was performed using HD-BET [14]. After that, FSL’s FAST was used to create the white matter mask [15]. DTI data were corrected for eddy currents and head motion with FSL’s eddy_correct. Diffusion parameters were computed with FSL’s DTIFIT. The principal eigenvector of the diffusion tensor was used to calculate the nerve fiber angle.
R2* was computed via a voxel-wise two-parameter mono-exponential single decay fit in MATLAB (The MathWorks, Inc., Natick, MA, United States), see equation (1) [3,4,16–19].
R_2,1^*(θ)=a_0+a_1*cos(θ)^2+a_2*cos(θ)^4 (1)
Fiber orientations were divided into 18 intervals of 5° each, covering the range from 0° (parallel to B0) to 90° (perpendicular to B0). Additionally, the data across subjects and groups were normalized to ensure comparability. All data underwent a linear registration to ensure the same pixel dimensions and orientation. Figure 1 displays the normalized R2* values, while Figure 2 presents the absolute R2* values as a function of the fiber angle for the three examined groups. Each graph represents the group’s mean values and standard deviations. The in-vivo group exhibited the most pronounced orientation-dependent variation with normalized R2* values ranging from 0.89 - 1.12. In the in-situ group, relative values ranged from 0.91 - 1.05, while relative values ranged from 0.94 - 1.01 for ex-situ cases. As expected, absolute R2* values were highest in the ex-situ group and lowest in the in-vivo group. Our results confirm the orientation dependency of R2* in all investigated conditions: in-vivo, in-situ and ex-situ. The strongest effects were observed in the in-vivo group, with deviations exceeding ± 10 %. In-situ and ex-situ conditions showed smaller effects. The three examined groups were scanned at different temperatures: in-vivo at the highest, ex-situ at intermediate room temperature and in-situ at the lowest temperatures. Since the in-situ condition resulted in an intermediate range of R2* values, it is unlikely that the observed differences in orientation dependency between groups are primarily due to temperature. This supports the hypothesis that formalin fixation influenced the observed R2* orientation dependency by altering tissue microstructure through protein cross-linking and dehydration, thereby reducing magnetic susceptibility differences and local field inhomogeneities [20–22]. Additionally, it is unlikely that the observed effect is attributable to ALS-related changes, as no differences were detected in the in-situ orientation dependency between ALS and neurologically healthy individuals. Our findings demonstrate an orientation sensitivity of R2*, with the lowest orientation dependency observed ex-situ. This suggests that formalin fixation alters tissue microstructure and reduces R2* orientation dependency, emphasizing the need for caution when interpreting ex-situ data from fixed brain tissue.
Lennart BEDARF (Basel, Switzerland), Dominique NEUHAUS, Maria Janina WENDEBOURG, Regina SCHLAEGER, Christoph BIRKL, Eva SCHEURER, Claudia LENZ
13:30 - 15:00
#47567 - PG254 Feasibility and Potential of Quantitative Susceptibility Mapping in Neurodegeneration with Brain Iron Accumulation (NBIA) at Clinical Magnetic Field.
PG254 Feasibility and Potential of Quantitative Susceptibility Mapping in Neurodegeneration with Brain Iron Accumulation (NBIA) at Clinical Magnetic Field.
Neurodegeneration with brain iron accumulation (NBIA) is a group of extremely rare diseases characterized by intracerebral iron accumulation and progressive neurodegeneration. To date, 11 genes have been implicated in NBIA [1]. The pathophysiology of NBIA is partially understood, although several pathways have already been identified, including mitochondrial homeostasis, lipid peroxidation, autophagy and iron metabolism. Although some imaging patterns suggest a specific NBIA subtype, there is no correlation between evolution of iron accumulation, stage of the disease and specific NBIA subtype. A quantitative measure of the iron load could then be used as a biomarker of the disease and for evaluating the pathology severity. Quantitative Susceptibility Mapping (QSM) provides an iron-load quantitative metric [2,3], with promising investigations in NBIA at ultra-high magnetic field [4,5]. Here we show feasibility in a clinical context at 3T.
MRI data were acquired at Bordeaux Hospital (France) on a General Electric 3 Tesla scanner with 48Rx head coil. A 3D gradient echo multi-echo sequence (SWAN) was applied following the consensus guidelines [6] (TA=4.8 min, FoV=224x224x176 mm, voxel size=1 mm isotropic, TR=34.7 ms, TE1=4.34 ms, ∆TE=5 ms, number of echoes=6, flip angle=16°, bandwidth=280 Hz/px, with compress-sensing) after a quality-control analysis on phantom and on a volunteer, the protocol was applied to NBIA patients. QSM reconstructions were performed in a secured-cloud European-based infrastructure[7]. A pipeline was implemented in Python, to compute R2* and QSM maps using MEDI [3,8], and extract values for several regions of interest (ROIs). Basal ganglia ROIs were segmented manually for each subject. Mean and standard deviations were computed in each ROI. We successfully implemented and validated an acquisition protocol for QSM in accordance with the QSM consensus guidelines, including the ability to use the compressed sensing option to reduce acquisition time without affecting phase for QSM on a clinical 3T system. Based on our quality-control procedures, with the applied protocol, we obtained on phantom a signal-to-noise ratio close to 225 with 1 mm isotropic acquisition providing QSM without significant bias (less than 1 ppb) and a standard deviation below 1 ppb. On the volunteer, average signal-to-noise ratio on the brain was larger than 100, indicating a field-mapping precision better than 1 ppb.
Patients analysed are three women, respectively affected by BPAN (linked to a mutation in the WDR45 gene), PKAN (PANK2), and MPAN (C19orf12). Increased iron accumulation in the basal ganglia was observed in NBIA, with a ~5-fold increase in pallidum on average, with values reaching up to more than 1000 ppb (Fig. 1), and a ~2-fold increase in caudate, putamen and substantia nigra on average over each ROIs. Interestingly, our results suggest a specific iron distribution profile in basal ganglia for each patient (Fig. 2), mainly in subtantia nigra and in pallidum. QSM give access to brain iron accumulation in NBIA patients on a clinical 3T system with clear individual iron accumulation profiles. These preliminary results indicate that QSM could be a useful biomarker to better understand the links between subtypes, affected regions, disease progression and iron accumulation. In the care pathway, it could be used for diagnosis and follow-up in rare diseases involving brain iron accumulation. QSM may provide a useful biomarker to evaluate efficiency and safety in the context of new therapy development.
Stephane ROCHE (Marseille), Samira MCHINDA, Anis BENYAHIA, Ludovic DE ROCHEFORT, Cyril GOIZET, Patricia FERGELOT MAURIN, Thomas TOURDAIS
13:30 - 15:00
#47927 - PG255 Can qMRI Be Utilized For Diagnosis of Parkinson's Disease at 7T: A Data Driven Approach.
PG255 Can qMRI Be Utilized For Diagnosis of Parkinson's Disease at 7T: A Data Driven Approach.
Automatic brain segmentation offers a scalable and objective alternative to manual methods, enabling the extraction of neuroanatomical features that may otherwise be impractical to assess. Given that patients with Parkinson’s disease (PDP) and healthy controls (HC) can be distinguished with MRI scans [1,2], we explore whether quantitative metrics derived from relevant regions of interest (ROIs) can effectively classify these two groups in an ongoing PD study [3].
45 subjects were included in this study; 24 patients already diagnosed with Parkinson’s disease (13 male, 11 female, mean age 65.8 years (SD=6.7)), and 21 age-matched healthy controls (10 male, 11 female, mean age 60.2 (SD=9.2)).
All MRI data were acquired using a Siemens Magnetom Terra 7T MR system (Siemens Healthineers, Erlangen, Germany) equipped with a 1TX/32RXhead coil (Nova Medical Inc, Wilmington, Delaware). The protocol included MP2RAGE [4] and a multi-echo GRE for QSM (ASPIRE sequence [5]). The resolution for both images was (0.75mm)³. TGV was used for QSM-processing [6].
ROIs that are important for Parkinson’s disease do not all have contrast on T1w images, so we used two pipelines for obtaining ROIs. For putamen, caudate, thalamus, hippocampus, amygdala and lateral ventricles we used FastSurferVINN (FSV) [7,8,9]. For substantia nigra (SN), red nucleus (RN) and subthalamic nucleus (STN) we used nnUNet [10] to train a UNet that segments on QSM.
For FSV, we first removed the background noise outside the skull of the MP2RAGE UNI image. We then ran FSV on it. The pipeline is illustrated in Figure 1, and Figure 2 shows an example of the segmentation.
The UNet was trained on 15 manual delineations from an independent, young and healthy population, and one manually delineated dataset from STRAT-PARK. It was trained to segment the SN, RN and STN. We used 5-fold cross-validation for training, as the training data was limited. The pipeline is illustrated in Figure 1, and Figure 2 shows an example of the segmentation.
The ROIs were coregistered, using Nighres [11], to the space of the other contrast to retrieve the map-values.
We used SPM12 to obtain total intracranial volumes (gray matter, white matter and CSF) to normalize volumes of the ROIs [12]. The pipeline is illustrated in Figure 1.
From the MP2RAGE and GRE sequences we obtained an R1-map, R2*-map and QSM, utilizing Nighres. For each map we extracted the mean, standard deviation, median, max, min, 5-percentile, skewness and kurtosis for each ROI. For each subject, the statistic values and volumes gave a vector with 225 features.
Before inputting the vectors into the classifiers, we scaled each feature using Z-score. We applied various classifiers: support vector machine (SVM), random forest, logistic regression (logreg), XGBoost and K-Nearest-Neighbours (knn) [13, 14]. For each classifier we performed a grid search to optimize hyperparameters. Even with a variety of classifiers, statistics from R1, R2* and QSM values for the ROIs we selected do not provide enough information to classify subjects as PDP or HC. The accuracies of each classifier are presented in Table 1.
For selected ROIs, statistics and contrast, the histograms of mean, 5-percentile, skewness, kurtosis and Jarque-Bera (measure of normality) are presented in Figure 1. Previous results have found a significant increase in the mean QSM value of STN, which is not detected in our cohort. Interestingly, the STN QSM-value in the PD group is similar in the two cohorts, while for the control group it is higher in our study compared to ref [2]. There seems to be bigger intra-class variation than inter-class variation in the two groups. On a group level we know that the putamen volume is significantly lower for PDP than for HC, but this does not show discriminative power.
This means that further work should consider other places in the brain than these ROIs to see if more information about the disease can be found. PD is, however, a complex disease and may actually need more complex analysis. Contrary to previous findings, for our cohort, this combination of statistics, ROIs and contrasts do not contain enough information to predict disease.
Anne Louise KRISTOFFERSEN (Trondheim, Norway), Runa Geirmundsdatter UNSGÅRD, Marc-Antoine FORTIN, Ingrid Gylterud KVÅLSGARD, Kjersti Eline STIGE, Thanh DOAN, Erik Magnus BERNTSEN, Charalampos TZOULIS, Pål Erik GOA
13:30 - 15:00
#47720 - PG256 CEST MRI for glycogen imaging in ex-vivo rat liver at 9.4T: A feasibility study.
PG256 CEST MRI for glycogen imaging in ex-vivo rat liver at 9.4T: A feasibility study.
Glycogen, a branched glucose polymer, serves as a short-term energy reserve and is primarily stored in the liver and muscles [1]. Disruptions in its metabolism can lead to metabolic pathologies that primarily affect these tissues.
In this context, Chemical Exchange Saturation Transfer (CEST) MRI has emerged as a promising non-invasive technique for detecting specific metabolites, including glycogen, in biological tissues [2]. GlycoCEST, which targets hydroxyl group protons in glycogen molecules, has already been applied to liver [3], which has a high glycogen content but also other organs such as brain [4] and skeletal muscles [2]. These studies have primarily focused on in vivo imaging in both healthy and pathological conditions.
In this study, we investigate the application of CEST imaging to ex vivo fixed liver tissue at 9.4 T. We begin by optimizing acquisition parameters using glycogen phantoms and subsequently demonstrate the feasibility of glycogen detection in ex vivo rat liver sample and validate it by colorimetric glycogen measurement.
In-vitro experiment: Three tubes containing 0, 50, and 200 mM of bovine glycogen dissolved in phosphate buffered saline (PBS) were prepared for in-vitro validation. To minimize frequency shifts between the phantoms, they were placed inside a larger plastic container filled with Thermasolv CF2 (Fig 1-a).
Ex-vivo experiment: Liver tissue (~2.6 g) was harvested from a 250 g, ~12-week-old female Wistar rat immediately after sacrifice. The liver was divided into different portions for complementary analyses. One portion was fixed in formaldehyde to prevent post-mortem physiological changes. During the experiment, the sample was placed in a syringe with Thermasolv (Fig 1-b) and maintained at 37 °C using a temperature-controlled water bath, with temperature monitored via a probe.
Colorimetric Quantification of Glycogen: The second portion of the liver was frozen in liquid nitrogen, then crushed prior to extraction. The procedure adapted from Schaubroeck et al. [6], involves alkaline digestion, ethanol precipitation, and resuspension in water. Quantification was performed using a commercial colorimetric assay kit (ab65620; Abcam) based on glycogen hydrolysis and enzymatic detection, with absorbance measured at 570 nm using a microplate reader.
MR acquisition & post-processing: Experiments were conducted on a 9.4T/30cm (Bruker Biospin MRI, Ettlingen, Germany) with a cylindrical transmit coil (87-mm intern) coupled with a 4-channel phased-array receive coil. A CEST FLASH sequence with continuous wave saturation was used, with frequency offsets from +3 to –3 ppm (0.1 ppm steps). For ex vivo sample, signal averaging was performed to improve signal-to noise ratio. To optimize acquisition parameters, various B₁ amplitudes and saturation durations were tested, as detailed in Table 1.
Initially, the CEST images were reconstructed, corrected for B₀ inhomogeneities using the Water Saturation Shift Referencing (WASSR) method [6] with offsets from +0.6 to –0.6 ppm (0.05 ppm steps) and data were fitted using a two pool Lorentzian fitting approach [7], implemented in MATLAB. After correction, Z-spectra were generated and Magnetization Transfer Ratio asymmetry MTRasym(Δω)= (S(-Δω)- S(Δω))/(S₀) was computed to highlight glycogen presence through its asymmetric CEST effect.
MTRasym at 0.7 ppm, corresponding to the glycogen peak [2-4], was then determined using manual ROIs in each phantoms and over the whole visible tissue in ex vivo liver (Fig 1-c,d). Figure 2 shows the Z-spectra for phantoms and rat liver at saturation time at 1 s and different B1 peak amplitudes. An asymmetry in the Z-spectra, particularly around the glycogen peak in the range of 0.5–1.5 ppm, is observed and increases with glycogen concentration.
According to table 2 which summarizes MTRasym values at 0.7 ppm, optimal CEST imaging was obtained with a B1 amplitude of 3 µT and 1.5 s saturation for in vitro experiments, and 2 µT with 1 s saturation for ex vivo sample, both yielding the max glycogen peak with MTRasym of 10.83 % (phantom of 200 mM) and 7.04 % at 0.7 ppm, respectively.
Colorimetric quantification revealed a glycogen concentration of 58.95 glucose units per mg of liver tissue, corresponding to 347 mM in glucose units. CEST imaging has rarely been applied to ex vivo samples [8-9]. Here, we tested and validated the sequence on glycogen phantoms, then tested them on ex vivo rat liver tissue. The observed CEST effect on both experiments matched the glycogen peak reported in in vivo glycoCEST studies (0.5–1.5 ppm) and post-mortem colorimetric analysis confirmed these findings, supporting the feasibility of ex vivo glycoCEST. These initial experiments serve as a foundational step toward validating the technique before its application to tissues with lower glycogen content, such as skeletal muscle and cardiac tissues. This will support future applications on metabolic alterations in arrhythmias and storage disorders.
Mathilde DEMORISE (Bordeaux), Julien FLAMENT, Fanny VAILLANT, Emma ABELL, Marion CONSTANTIN, Nestor PALLARES-LUPON, Richard WALTON, Bruno QUESSON, Olivier BERNUS, Julie MAGAT, Arash FORODIGHASEMABADI
13:30 - 15:00
#47620 - PG257 Tracking Remote Cervical Cord Atrophy in Degenerative Cervical Myelopathy after Treatment: A Large Cohort Longitudinal MRI Study.
PG257 Tracking Remote Cervical Cord Atrophy in Degenerative Cervical Myelopathy after Treatment: A Large Cohort Longitudinal MRI Study.
Degenerative cervical myelopathy (DCM) is the most common form of non-traumatic spinal cord injury. Cord tissue damage is mainly attributed to cervical compression, while other factors are understudied. Treatment decisions in DCM are challenging due to variable disease progression and a lack of reliable biomarkers[1,2]. Longitudinal structural MRI can provide insight into the temporal pattern of subsequent remote structural tissue changes. This study aims to investigate long-term tissue atrophy using structural T2*-weighted MRI in the cervical cord rostral to the compression site in patients who underwent surgical decompression (operated (OP)) or received conventional non-surgical treatment (non-operated (nOP)).
Data acquisition
In this study, 102 DCM patients (mean age±SD: 55.74±11.42 years, 61 males) underwent longitudinal MRI scans of the cervical cord at C3 using 3T MR scanner (MAGNETOM Skyra Fit or Prisma; Siemens Healthineers) equipped with a 20-channel head/neck RF coil. The MRI protocol consisted of sagittal TSE T2- and axial T2*-weighted MRI and clinical examinations at three time points: baseline, 6-, and 12-month follow-ups. Clinical protocol consisted of the International Standards for Neurological Classification of Spinal Cord Injury protocol[3] for evaluation of cervical light-touch and pin-prick scores and total modified Japanese Orthopaedic Association (mJOA) scale[4]. Forty-one DCM patients were operated following baseline clinical assessment. Seventy-two DCM were classified as mild (15≤mJOA≤17) and 30 as moderate DCM (mJOA≤14) at baseline. For comparison, structural MRI data were acquired in 10 healthy controls (HC) (mean age±SD: 45.30±18.54 years, 7 males) at 0, 6, and 12 months.
Image processing
The cross-sectional area (CSA) of the spinal cord (SCA), grey matter (GMA) and dorsal columns (DCA) were derived from manual segmentations obtained using JIM software (v9). The CSA was averaged across slices covering C3 level. White matter area (WMA) was calculated from subtraction of GMA from SCA[5].
Statistical analysis
To ensure greater clinical homogeneity of the compared groups, statistical analysis was conducted in the mild and moderate DCM cohorts separately. For each, two-tailed Fisher’s exact tests were conducted to assess differences in clinical scores at baseline between OP and nOP. An ANOVA was performed to investigate WMA, GMA, and DCA differences between OP, nOP, and HC. Linear models with age as covariate were conducted to determine longitudinal rate of change of clinical scores and tissue-specific CSA, in OP and nOP. At baseline, WMA, GMA, and DCA derived from T2*-weighted MRI were comparable between OP and nOP groups in both mild and moderate DCM (Fig1). Similarly, clinical scores did not differ between groups in either severity category (Fig2).
Both treatment groups (OP and nOP) showed a significant GMA decrease over time, in mild (p = 0.0003) and moderate DCM (p = 0.03) (Fig3).
In mild DCM, OP patients showed significant clinical improvement over 12 months post-surgery (Fig4a), while nOP patients showed no significant change. In moderate DCM, both OP and nOP patients improved in mJOA score over 12 months (Fig4b). Although mild and moderate DCM patients improved in clinical assessment following decompression surgery, MRI revealed DCM-induced long-term grey matter atrophy remote from the compression site in both operated and non-operated patients with mild and moderate DCM. Interestingly, similar patterns of ongoing atrophy have been reported in acute spinal cord injury patients[6,7]. The current findings suggest that, even after successful decompression in DCM, secondary processes may continue to drive atrophy offering insight into DCM progression. Remote grey matter changes rostral the compression site, as observed on structural MRI, may result from anterograde and retrograde degeneration of sensorimotor spinal pathways[8]. The cross-sectional area remains a relatively coarse marker of neurodegeneration and should be interpreted alongside other DCM-specific tools and quantitative MRI sensitive to microstructural changes (e.g., DTI, T1 relaxometry) for a more comprehensive understanding of DCM progression. There was clinical improvement in patients with mild and moderate DCM following decompressive surgery, yet grey matter atrophy at C3 level was progressive, suggestive of continuing structural degeneration in this cohort. The clinical significance of these findings still needs to be determined, which can be facilitated through longer follow-ups and a comparison to more severely affected individuals. The study’s methodology – combining MRI with clinical assessment across multiple time points – establishes a framework aiming at reaching a more robust understanding of the natural and postoperative course of DCM. Future research will focus on investigating further time points (e.g., 2-year, 5-year follow-ups) to shed light on the long-term evolution of clinical symptoms and atrophy rate.
Anna LEBRET (Zurich, Switzerland), Markus HUPP, Nikolai PFENDER, Carl ZIPSER, Armin CURT, Patrick FREUND, Maryam SEIF
13:30 - 15:00
#47629 - PG258 Assessing multi-center reproducibility of APTw-CEST MRI in the healthy brain at 3T.
PG258 Assessing multi-center reproducibility of APTw-CEST MRI in the healthy brain at 3T.
The potential of amide proton transfer weighted chemical exchange saturation transfer imaging (APTw-CEST) in neuro-oncology has been widely demonstrated [1-3]. Still, clinical implementation remains challenging due to a lack of standardization. Consensus recommendations on data acquisition and analysis for APTw-CEST have been published for brain tumor imaging [4], and reproducibility and repeatability have been proven in various single-site, single-vendor studies [5-8]. Recently, we reported a variability of 12%-57% in APTw-CEST MRI acquired using different scanners and protocols in a phantom [9]. To investigate the ability to distinguish healthy and brain tumour tissue across sites and vendors, we have conducted the first in vivo, multi-site, multi-vendor reproducibility study of APTw-CEST MRI.
Two healthy volunteers were scanned using 3T MRIs at four different sites in the Netherlands that have MRI scanners of three different vendors: Amsterdam UMC (AUMC): Siemens (Vida, Siemens Healthineers, Erlangen, Germany); Erasmus MC Rotterdam (EMC): GE (Premier, General Electric, Chicago, USA); UMC Utrecht (UMCU), Leiden UMC (LUMC): Philips (Ingenia Elition X, Philips Healthcare, Best, The Netherlands). Written informed consent was obtained from both volunteers under local ethical approval.
A standard high-resolution T1w image was acquired as part of the scanning protocol at all sites. The acquisition details of the ATPw-CEST sequences are presented in Table 1. The acquisition protocols for AUMC and EMC were chosen based on the consensus recommendations [4]. Acquisitions at LUMC and UMCU were done using the 3D APT product sequence of Philips.
Magnetization transfer ratio asymmetry (MTRasym) was calculated using on-scanner processing software for the Philips datasets (LUMC and UMCU) and using in-house processing pipelines in Matlab and the FMRIB Software Library (FSL 5.0, Oxford, UK) for the Siemens (AUMC) [5] and GE (EMC) datasets [6].
Regions of interest (ROIs) were selected using FAST from FSL. The segmented ROIs were white matter (WM) and grey matter (GM). ROI averages and standard deviation (SD) of the MTRasym were calculated per volunteer per ROI. The between-scanner coefficient of variation (CoV) was calculated for each ROI per volunteer by dividing the mean MTRasym of the four scans by the standard deviation of the four scans. APTw CEST MRI values in both healthy volunteers were low and consistent with expected normative ranges for each acquisition, as seen in the example images in Figure 1. Mean MTRasym signal intensities were between -0.133±1.95 and 0.519±1.71 in GM and between 0.048±0.28 and 0.659±1.36 in WM (Figure 2, Table 2). The between-scanner CoVs range from 40% to 258% across the ROIs and volunteers (Table 2). The MTRasym values between -0.133% and 0.659% align with reported values for healthy brain tissue [4,5,6]. These values are lower than values for MTRasym in high-grade glioma (HGG), which are reported around 1.6% in the literature [5,6,10]. There is minimal overlap between the SDs of the healthy brain reported here and HGG (from literature), and the range of signal values expected in tumor tissue lies outside the standard deviation of the APTw-CEST values found in three of the four centers in the healthy tissue. The EMC MTRasym values overlap most with the HGG values and differ most from the other three sites. This, in combination with the low intensities measured in this study, has resulted in the high CoVs. Future work will focus on the standardization of post-processing to harmonize MTRasym across the four sites, which is expected to reduce the SD (both within an ROI, as well as between sites), further harmonizing the resulting APTw-CEST quantification.
A larger number of volunteers would have improved estimation of the measurement accuracy and variability; however, it would not have allowed us to interpret more from the results, due to the lack of APTw-CEST contrast in the healthy brain. Instead, we have initiated a large, multi-center patient study in which 120 patients with HGG will be recruited across the four sites to give further insight into the in vivo reproducibility of APTw-CEST MRI in the presence of contrast-enhancing pathology. Combined with the results from the previous phantom study, we hypothesize that, while currently MTRasym differs between sites and scanners, a standardized threshold MTRasym value for diagnosing HGG is likely to be attainable using these acquisition protocols.
Laura KEMPER (Rotterdam, The Netherlands), Chloe NAJAC, Joost KUIJER, Matthias VAN OSCH, Elsmarieke VAN DE GIESSEN, Marion SMITS, Evita WIEGERS, Esther WARNERT
13:30 - 15:00
#46879 - PG259 Cardiac MR-Based Assessment of Myocardial Viability Before Transplantation: A Porcine Feasibility Study.
PG259 Cardiac MR-Based Assessment of Myocardial Viability Before Transplantation: A Porcine Feasibility Study.
Accurate assessment of myocardial viability is essential to reduce the risk of early graft dysfunction following cardiac transplantation. Cardiac magnetic resonance (CMR) is a non-invasive reference technique for characterizing myocardial structure and composition, including edema, fibrosis, and energy metabolism. This study aimed to develop a fast and efficient imaging protocol for tracking myocardial viability over time, potentially enabling surgeons to better evaluate graft quality before transplantation.
Eight hearts were harvested from healthy pigs. Immediately after excision, cardioplegia was administered to arrest cardiac activity and preserve myocardial tissue. Following this, each heart was carefully placed in a container filled with cardioplegic solution to maintain tissue viability. To ensure consistent positioning, the hearts were stabilized inside the container. Imaging was performed using a 1.5T scanner (Siemens Healthineers, Germany), equipped with a 18-channel surface coil. For ¹H-MRS, a PRESS (Point-Resolved Spectroscopy) sequence was used. A single voxel was placed in the interventricular septum. Voxel placement was guided by anatomical localization on four-chamber and short-axis MRI views. Spectroscopic acquisition parameters were: TE=135ms, TR=1500ms, 1024 acquisition points per spectrum, and a spectral bandwidth of 1000Hz. Two types of spectra were acquired: 1) Water-suppressed spectra (32-512 NEX) were used to quantify intramyocardial metabolites, 2) Non-water-suppressed spectra (4-16 NEX) served as references for metabolite quantification. In parallel, T1 mapping of the myocardium was performed using a MOLLI sequence. Imaging parameters were: FOV=150×175mm², a matrix size of 110×128 (determining in-plane resolution), a slice thickness of 8 mm, and TE/TR of 1.21ms and 2.3ms, respectively. Both ¹H-MRS and T1 mapping acquisitions were repeated every hour for a total of 4 hours to assess temporal changes in myocardial viability markers. Spectral data were processed using dedicated post-processing software4. In the resulting spectra, a peak at 1.3ppm5 was interpreted as a composite signal from lactate and lipids, which typically increase in necrotic tissue. Quantification was performed relative to the water signal, and results were expressed as percent concentrations. For T1 analysis, the mid-ventricular short-axis slice was manually contoured once to delineate the endocardial and epicardial borders, and these same contours were propagated across all time points to ensure consistent region-of-interest analysis. The extracted T1 values provided a surrogate marker of tissue integrity and edema, which typically increase with ischemic injury. Quantification of intramyocardial metabolites using ¹H-MRS was successful in 6 out of the 8 hearts, representing 75% of the dataset. The remaining two hearts were excluded from metabolic analysis due to insufficient SNR in the acquired spectra. An example of a high-quality water-suppressed ¹H-MRS spectrum, acquired one hour after heart explantation, is presented in Figure1. The spectrum clearly shows distinct resonance peaks, particularly around 1.3ppm, corresponding to lactate/lipids. The mean relative concentrations (expressed as percentages of the water signal) of lactate and lipids combined were measured at four different time points post-explantation: Hour 1: 0.024±0.012%, Hour 2: 0.033±0.006%, Hour 3: 0.033±0.010% and Hour 4: 0.037±0.013%. These results show a gradual increase in signal intensity at 1.3ppm over time, which is likely driven by rising lactate concentrations which is a known marker of anaerobic metabolism, suggesting that ischemic processes continue to develop even under cold cardioplegic preservation. In contrast, the lipid component of this peak is expected to remain relatively stable, so the observed increase is interpreted as a surrogate for progressive metabolic distress. In parallel, T1 mapping data were acquired in all hearts. The T1 maps showed spatial homogeneity across the myocardium at each time point, indicating uniform relaxation properties (Figure 2A). The mean native myocardial T1 values increased over time as follows: Hour 1: 727±94ms, Hour 4: 765±89ms. This gradual increase in T1 relaxation time is consistent with the onset of ischemic injury, as T1 values are known to rise in response to edema formation, loss of membrane integrity, and cellular degradation. These findings are illustrated in Figure 2B, which summarizes the temporal progression of T1 values. This study introduces a combined ¹H-MRS and T1 mapping protocol to monitor myocardial viability in an ex vivo pig heart model. The observed parallel increase in lactate signal and T1 values over time likely reflects ongoing ischemic injury. We hypothesize that beyond a certain threshold—yet to be defined—myocardial viability may become irreversibly compromised. Further studies with larger cohorts are needed before clinical application in human heart graft assessment.
Khaoula BOUAZIZI (Paris), Elodie BERG, Noémie LACOURT, Mohamed ZARAI, Francesca BRANZOLI, Solenn TOUPIN, Yann LE FUR, Monique BERNARD, Guillaume LEBRETON, Alban REDHEUIL, Nadjia KACHENOURA, Frank KOBER
13:30 - 15:00
#47758 - PG260 Establishing In Vivo DTI for Cognitive Behavior in Wild Birds: Egg Rejection in Magpies.
PG260 Establishing In Vivo DTI for Cognitive Behavior in Wild Birds: Egg Rejection in Magpies.
Egg discrimination and rejection is the primary defensive mechanism used by hosts of brood parasites to counteract the negative effects of parasitism, since, in most cases, parasitized hosts would otherwise raise only parasitic offspring, leading to a complete loss of their own reproductive success (1). However, egg rejection occurs at relatively low frequencies. One proposed mechanism underlying discrimination is prolonged learning of the hosts’ own egg appearance, allowing them to recognize and reject foreign eggs (2). This learning process would involve the creation and memorization of a template of their own eggs, which could be used to identify mismatched eggs across multiple breeding attempts. It has been demonstrated that the hippocampus is the brain region responsible for memory, while the nidopallium is implicated in learning processes (3). The aim of this study was, for the first time, to compare these two brain regions between egg-rejecting and egg-accepting females using in vivo Diffusion Tensor Imaging (DTI) and assess anatomical brain volume changes between sexes.
Wild magpies were trapped in the town of Calahorra, Granada, Spain. Then they were moved to the preclinical MRI facility of the University, where they were anesthetized with 2% isoflurane in oxygen via a veterinary mask for induction and maintained under 1–2% isoflurane using a nose cone during scanning. Animals were positioned supine, restrained with tape, and placed over a water-heated blanket. MRI scans were acquired on a 7T Bruker Biospec scanner (20 cm bore, Paravision 360 v3.5) using a 1H rat brain surface coil and an actively detuned transmit-only resonator. Anatomical images were obtained with a variable flip angle 3D-T2-weighted RARE for brain volume assessment. For diffusion MRI, field mapping were performed prior to acquisition. DTI data were acquired using a two-shot EPI sequence with 60 diffusion directions (b=1000 s/mm²) and 10 b0 images. (TR/TE=7000/25 ms, δ/Δ=4/12 ms), total study time ~75 min (figure 1). 3D-T2WI were denoised employing 3D adaptative multiresolution nonlocal means denoising algorithm (4) and DWIs were processed using mrtrix3 software (5). Regions of interest involved in learning, problem solving, and memory (hippocampus, mesopallium, nidopallium, and tectum opticum) were delineated blindly and manually by a an avian neurobiologist, and mean value of the ROI was employed in statistical t-test. Preliminary data point that females (n=6) exhibited larger brains than males, even after normalizing for body weight (figure 2), suggesting potential sex-based differences in neuroanatomical organization (6). Preliminary significant differences in DTI-derived parameters (including fractional anisotropy (FA), mean diffusivity (MD), and axial diffusivity (AD)) were found between females that rejected parasitic eggs and those that did not (n=4 per group). Specifically, higher FA, MD, and AD values were observed in non-rejecting females, which may reflect differences in hippocampal microstructure (figure 3). These findings, together with the hippocampal differences observed between groups (and the absence of differences in the nidopallium), suggest that the ability to discriminate between own and parasitic eggs could be linked to local neuro-architectural variations. One plausible explanation is a greater organization of interneurons in the hippocampus of rejecting females, supporting enhanced processing capabilities related to egg recognition. Previous studies have shown that DTI metrics can reflect underlying neuronal composition in avian brain (6,7), supporting the hypothesis that rejecting females may exhibit a more refined brain microstructure. Although further behavioral recorded video-data are needed to be analyzed to confirm this structure-function relationship, our findings point toward a neurobiological basis for individual variation in parasitic egg discrimination. We successfully established an in vivo MRI and DTI protocol for wild magpies, enabling the investigation of neuroanatomical correlations of egg rejection behavior. Our results reveal significant microstructural differences in the hippocampus between egg-rejecting and non-rejecting females, suggesting a potential neurobiological basis for individual variation in parasite egg discrimination.
Balbino YAGUE (Granada, Spain), Juan Gabriel MARTÍNEZ, Mercedes MOLINA
13:30 - 15:00
#47569 - PG261 Evaluation of an Acceleration Method for Mouse Brain Diffusion Tensor Imaging.
PG261 Evaluation of an Acceleration Method for Mouse Brain Diffusion Tensor Imaging.
Diffusion is a key technique in Magnetic Resonance Imaging for probing the brain’s microstructural organization. It provides critical biomarkers such as fractional anisotropy (FA), or tractography, which enables the reconstruction of structural connectivity between brain regions. These analyses typically rely on Diffusion Tensor Imaging (DTI) sequences.
To achieve high-resolution 3D maps in small animals and employ advanced models such as High Angular Resolution Diffusion Imaging, a large number of diffusion gradient directions must be acquired. This requirement leads to prolonged acquisition times—often exceeding 10 hours—thus precluding in-vivo applications.
To address this limitation, one strategy is to reduce the number of acquired measurements and apply Compressed Sensing reconstruction techniques to recover relevant diffusion contrasts.
Among these, low-rank reconstruction methods exploit the redundancy across neighboring voxels by assuming similar signal behavior within local spatial patches, without requiring explicit signal models. Alternatively, subspace reconstruction methods incorporate prior knowledge of signal behavior through simulations, enabling data recovery by projecting onto a learned signal subspace.
This study proposes a proof of concept demonstrating that the combination of an advanced reconstruction framework with optimized and original sampling schemes enable substantial acceleration of 3D diffusion MRI acquisitions of the mouse brain while preserving image quality and the reliability of derived diffusion metrics.
Materials:
Experiments were performed on a 7T Bruker BioSpec system (Ettlingen, Germany) equipped with a volume resonator for excitation, and a 4-element (2×2) phased surface cryoprobe for signal reception.
Sequence:
A 3D Spin-Echo sequence was applied with a 156µm isotropic resolution, matrix: 128x128x96, 30 directions, 5 b0, total acquisition time=12h. Ex vivo healthy-mouse brains were used, after brain extraction and PFA fixation.
Acceleration:
Acceleration was performed by a posteriori removing data based on a Variable-density Poisson Law along the phase and partition axes. The center of the 3D k-space was left fully encoded. A random sampling was performed along the diffusion directions. Different acceleration factors (from 6 to 12) were tested.
Reconstruction:
For reconstruction we used two methods: Low Rank reconstruction and Subspace reconstruction:
- For Low Rank reconstruction we used the Local Low Rank (LLR) with a wavelet regularization. We optimise both reguliization parameters for low rank and wavelet
- For the subspace reconstruction, two dictionnaries were built. One based on simulation based on the DTI model by creating a large number of ellipsoids with different eigen values and different orientations. The other is based on the fully sampled central data, by reconstruct low resolution images.
The simulation and reconstruction have been perfomed using Julia and the package MRIReco.jl and the Bart ToolBox (Berkeley Advanced Reconstruction Tool)
Image analysis:
FA maps were reconstructed using Dipy on Python to validate the acceleration process by comparing the standard deviation value between undersampled and fully sampled data as a reference. For the low-rank reconstruction, an ℓ1-regularization was applied in the wavelet domain along the three spatial dimensions, while a Locally Low-Rank regularization was performed along the diffusion dimension. The regularization parameters were optimized by minimizing the Mean Squared Error between the accelerated acquisition and a fully sampled reference. (Fig.1)
For the subspace reconstruction : the optimal number of subspace dimensions was determined by testing various values on fully sampled data. The results indicated that, although the singular values decay rapidly, a relatively large number of components is required to ensure accurate reconstruction (>10). (Fig.2)
Comparison of the reconstructions from under-sampled data using both the subspace and low-rank approaches suggests that, under the current simulation setup, both methods yield comparable results even if subspace reconstruction tend to offer a more accurate reconstruction. Less than ± 1% error has been calculated beetwen the mean FA value of the fully sampled corpusm callux (0.488 ± 0.097) and an acceleration factor 6 subspace reconstruction with a simulated dictionnary (0.489 ± 0.095). The Low Rank reconstruction with the same acceleration factor shows a difference of 5%. An acceleration factor of 6 can be applied without compromising image quality for both reconstruction approaches (Fig.3) corresponding to an acquisition time of approximately 2 hours.
FA maps confirmed that accelerating the acquisition by a factor of 6 did not alter the resulting diffusion metrics.
However, the current acquisition time still remains relatively long, and further optimization is necessary to validate the feasibility of using higher acceleration factors.
Nicolas SIMONNEAU (Bordeaux), Elise COSENZA, Emeline RIBOT, Laurent PETIT, Sylvain MIRAUX, Aurélien TROTIER
13:30 - 15:00
#46509 - PG262 A subvoxel correction of boiling-induced susceptibility artifacts in magnetic resonance thermometry and dosimetry for monitoring microwave thermoablation.
PG262 A subvoxel correction of boiling-induced susceptibility artifacts in magnetic resonance thermometry and dosimetry for monitoring microwave thermoablation.
Real-time monitoring of microwave liver ablation (MWA) using MRI thermometry can be hindered by susceptibility artefacts [1,2] caused by boiling [3]. The latter provoke unphysical temperature deviations that prevent lesion size prediction [4,5,6]. This study proposes a correction based on removing the contribution of the susceptibility artifact using subvoxel sources of susceptibility.
Experiments: The protocol was approved by the ethics committee for animal experimentation CEEA50 (France). MWA (N=23) were performed using an AveCure microwave system (14-gauge large antenna, MedWave, USA) on seven pig livers. Energy delivery was set to 7’30 with a target temperature of 80°C.
Acquisitions and dosimetry: Acquisitions were performed under respiratory gating during the most stable part of exhalation using multi-shot gradient-echo echo planar imaging sequences on 1.5T and 3T Siemens. The voxel size was 2.3 x 2.3 x 3.0 mm3, slice gap 1.5 mm with 7 slices. At the end of the experiment, gadoteric acid (0.5 mmol/kg, GE Healthcare) was injected intravenously, and 3D T1-weighted images were acquired to visualize the non-perfused volumes at a resolution of 1.2x1.2x1.2 mm3. Temperature calculation was performed using the PRFS method, followed by a spatial-temporal drift correction. The proposed correction of susceptibility artifacts was then applied. The cumulative TD based on the Sapareto equation was computed, taking an equivalent dose of 240 min at 43°C (CEM43) as the lesion volume (‘TD ablation zone’)
Susceptibility correction methodology: Figure 1 shows a schematic overview of the method. It consists in generating a susceptibility distribution that reproduces the main stable component of the artifact through time. The magnetic susceptibility artifact simulation used the dipole approximation of the field perturbation caused by local changes of magnetic susceptibility. The susceptibility values were determined at a finer resolution (subvoxel) and using adjacent slices contribution by solving a linear inverse problem which was set to nullify the artifact.
Temperature data and lesion size analysis: The data analysis was performed retrospectively, aimed at assessing the ability of the method to reproduce the artifacts and correct the temperature deviations. Second, to demonstrate the advantages of this method under actual treatment conditions, comparisons were performed with the postprocedural ‘T1w ablation zone’ against two ‘TD ablation zones’: one given by the uncorrected data and one given by the proposed correction. Out of seven pigs with 23 MWA, 3 (13%) MR temperature acquisitions were excluded due to low signal-to-noise ratio (SNR). Susceptiblity artifacts caused by boiling were observed in 11 (48%) acquisitions.
Figure 2 presents a selection of corrections over six MWA with varying degrees of: artifact intensity, shapes of the artifact, orientations of the ablation probe in respect to the slice and B0 magnitudes (1.5T and 3T).
Figure 3 compares the time evolution of temperature and thermal dose for a slice adjacent to the MW probe that was impacted by a positive lobe of the artifact. The 10°C offsets visible in the marked voxels are removed after the correction, causing a considerable impact on the dosimetry estimate, as the TD threshold is no longer reached in those voxels.
Figure 4 displays the T1w ablation zone segmented from post-contrast images versus TD ablation zones with and without the correction. The first case shows a discontinuous lesion shape due to the presence of a vessel, while the other shows an ellipsoid-shaped lesion. Without correction, an overestimation of lesion size up to 9 mm (two slices of 4.5 mm thickness) in a minor axis was observed.
The geometric and overlap metrics indicate an increase of agreement between corrected-and-T1w measures versus uncorrected-and-T1w: (i) overestimation of lesion volume (T1w-TD) was reduced in average from -1.02 to -0.59 cm³ (42% reduction), most noticeable in Y (26% reduction in average from -3.46 to -2.55 mm) and Z axis (17% reduction in average from -3.20 to -2.67 mm); (ii) median volumetric Dice increased by 4.8% (0.62 to 0.65). The use of subvoxels was motivated to diminish the main pitfall in the dipole model: for positions close to the point of non-zero susceptibility, the kernel will induce deviations that grow without bounds. The prior selection is a potential limitation. A perfect correction would require a time dependent simulation. However, the artifact is relatively stable through time. Comparison with T1w imaging showed improvements in prediction of lesion volume. The proposed algorithm takes into account partial volume effects and the contribution of adjacent slices. It possesses the necessary adaptability for simulating a diverse range of deformed dipole-like artifacts seen in experimental data. It could also be applied to radiofrequency or laser energy.
Eber DANTAS DE SÁ PAIVA, Rebecca LAFONT, Pierre BOUR, Thibaut FALLER, Bruno QUESSON, Helcio ORLANDE, Valery OZENNE (Bordeaux)
13:30 - 15:00
#45842 - PG263 hMRI toolbox: a comprehensive tool for in vivo MRI histology with built-in denoising, error quantification and motion mitigation.
PG263 hMRI toolbox: a comprehensive tool for in vivo MRI histology with built-in denoising, error quantification and motion mitigation.
The hMRI toolbox is an SPM-based, collaboratively developed open source software on GitHub for generating quantitative MRI (qMRI) maps sensitive to myelin and iron content [1]. It produces high-quality multiple quantitative parameter maps (R1, R2*, PD, and MTsat) that facilitate advanced analyses, precise subcortical delineation, and provide critical inputs for in vivo histology via biophysical models [2]. Here we highlight the upcoming 1.0 release, containing updates to tissue weighted smoothing and mask creation as well as new capabilities such as advanced image denoising, the quantification of errors on the qMRI estimates, and a correction for motion-related image degradation in statistical analyses (QUIQI) [3-6]. These features seamlessly integrate with the existing DICOM import, map creation and qMRI map processing (spatial processing) modules (Fig.1). Furthermore, the toolbox can now run as a standalone application, is available in Neurodesk [7] and is supported by the upload of the public demo dataset [8,9] to GitHub by utilizing the Git “large file storage” (LFS) (Fig.2), promoting reproducible research and community engagement.
The updated hMRI toolbox has been re-engineered as a standalone application that can run on platforms such as Neurodesk, allowing users to operate the toolbox without relying on a MATLAB license. At the start of the processing workflow (Fig.1) the DICOM Import module facilitates the conversion of raw DICOM files to NIfTI format. Following this, a state-of-the-art denoising module [3]—integrated via a custom Java-MATLAB interface— provides access to a set of advanced noise reduction techniques [10-12] (Fig.3). Subsequently, the Create Maps module converts the preprocessed data into quantitative maps. The Error Maps module (Fig.4a), quantifies voxelwise errors on the qMRI estimates and can adaptively combine multiple sets of qMRI data to downweight regions with high error. In group-level analyses, the QUIQI module (Fig.4b) accounts for the level of motion-related degradation of each individual map to ensure the validity of the statistical results. The index of motion degradation estimated by the Create Maps module of the toolbox [13] or another index of the user’s choice can be used. Enhanced metadata support is provided through automatically generated JSON sidecar files, and the inclusion of a public demo dataset [8] (Fig.2) allows users to validate and refine the workflow. The processing workflow in hMRI toolbox 1.0 provides substantial improvements in both usability and data quality control. The standalone version successfully broadens accessibility by eliminating the dependency on MATLAB licensing, simplifying deployment across varied computational environments. Users can now employ a seamless framework beginning with the DICOM Import module, followed by advanced denoising, and subsequently the creation of quantitative maps via the Create Maps module (Fig.1). This integrated approach ensures that the output maps exhibit markedly reduced noise [3] (Fig.3). In addition, the Error Maps module provides voxelwise assessments of map quality that allow the adaptive combination of maps acquired across different repetitions. The QUIQI module allows the mitigation of motion-related image degradation to ensure the validity of statistical results in group-level analyses (Fig.4). Testing on the public demo dataset [8] confirmed that these new modules can enhance users’ capability to conduct analyses of microstructural brain tissue change from qMRI data. The recent enhancements in the hMRI toolbox epitomize a strategic refinement in the processing of quantitative MRI data. The addition of state-of-the-art denoising techniques, coupled with the detailed Error Maps module and the QUIQI module for motion compensation, further refine the map creation process. The ability to use these modules in a workflow—beginning with DICOM Import, progressing through denoising and map creation, with the option to apply error assessment, motion mitigation, and dedicated spatial processing—creates a cohesive processing tool that significantly enhances data quality and reproducibility. The standalone application expands accessibility to diverse computational environments like Neurodesk. hMRI toolbox 1.0 is a significant advancement in quantitative MRI analysis. By combining reliable DICOM import, map creation and spatial processing (Fig.1) with state-of-the-art denoising [3], meticulous error mapping (Fig.4a), and effective motion mitigation (Fig.4b), the toolbox delivers robust qMRI biomarkers critical for in vivo histology. Its utility spans a diverse range of applications—including developmental and aging studies [14,15] and explorations of neurodegenerative or neuroinflammatory conditions [16,17]—cementing its role as a powerful resource for enhancing research efficiency and fostering open source software development and reproducible neuroimaging methodologies.
Baris Evren UGURCAN (Leipzig, Germany), Christophe PHILLIPS, Luke EDWARDS, Antoine LUTTI, Martina F. CALLAGHAN, Siawoosh MOHAMMADI, Pierre-Louis BAZIN, Gunther HELMS, Karsten TABELOW, Nikolaus WEISKOPF
13:30 - 15:00
#46818 - PG263 MADI@700: A Submillimeter, Openly Available, and Replicable diffusion MRI Dataset of 9 In-Vivo Macaque Brains.
PG263 MADI@700: A Submillimeter, Openly Available, and Replicable diffusion MRI Dataset of 9 In-Vivo Macaque Brains.
Due to the phylogenetic proximity and the strong homology between the human and non-human primates’ brain, investigating the neuroanatomy of the macaque brain helps further our understanding of the human brain and the development of novel therapeutic approaches. Diffusion-weighted MRI is a powerful non-invasive tool to investigate the microstructure of the brain[1]; however, it remains challenging to acquire in vivo data in macaque monkeys. Their brain size requires higher spatial resolution, but their mensuration generally prevents the use of preclinical scanners designed with higher magnetic fields. Here, we propose a replicable method to acquire sub-millimeter isotropic diffusion MR imaging on anesthetized primates with a standard clinical MRI scanner. We also make available to the scientific community a dataset on 9 macaque brains comprising raw diffusion and anatomical MRI data, the associated templates, and the data curated for MR tractography.
We acquired data from 5 male and 4 female healthy macaque monkeys (Macaca fascicularis) aged between 3 and 23 years old at the time of experiments. All experiments were performed with a 3T Prisma Siemens MR scanner, using the vendor’s body coil for transmission and a 64-channel head coil for signal reception, as commonly used for human acquisitions. The animals were positioned in supine position, with their head maintained straight in the MRI coil using a shape memory head support (MOLDCARE head cushion). Each animal underwent two MRI sessions dedicated to diffusion imaging (2h36min) and anatomical imaging (1h11min). The animals were intubated and maintained under anesthesia using 1-3% isoflurane mixed with pure oxygen.
Diffusion-weighted acquisitions were obtained with an in-house MR pulse sequence, a 3D spin-echo segmented EPI, designed to maximize SNR and provide high isotropic resolution while maintaining the total acquisition time below 3 hours for the animal's wellness. Images were reconstructed using the standard vendor’s reconstruction. Key parameters were: Axial orientation, TR/TE= 700/64ms, receiver bandwidth= 776 Hz/pixel, Partial Fourier 7/8, 4 segments, FOV=100 x 102.9 x 61.6 mm3, nominal isotropic resolution of 0.7 mm3, 32 diffusion encoding directions at b=1000s/mm2, two acquisitions with b=0s/mm2 (named b0) and opposite phase encoding directions.
In addition, high-resolution T1-weighted (MPRAGE) and T2-weighted (SPACE) acquisitions with isotropic resolution of 0.5mm3 were collected in a separate session with Siemens product sequences.
All images were processed using versaFlow[2], an openly available pipeline designed to preprocess and analyze diffusion MRI data from macaque brains. Diffusion MRI preprocessing included filtering for background noise, magnetic susceptibility, eddy currents, motion, and intensity bias. Diffusion profiles were obtained by Diffusion Tensor Imaging (DTI)[3] and Constrained Spherical Deconvolution (CSD)[4].
Probabilistic local tractography[5], [6] was performed with a GPU implementation in the Sherbrooke Connectivity Imaging Lab (SCIL) Python dMRI processing toolbox (Scilpy) on the whole brain. The common template space was generated from all 9 subjects using a multi-modal registration approach available in ANTs. The parcellation of the INIA19 atlas[7] was used as regions of interest (ROIs) to extract some bundles of interest, namely the corpus callosum (CC), the anterior commissure (AC), and the pyramidal tract (PyT). Quality control assessment was performed for the 9 datasets at each step of the versaFlow pipeline, using the Diffusion MRI Quality Check in the Python (dmriQCpy) toolbox[8]. T1, T2 and diffusion weighted images acquired for each of the 9 animals are shown in Figure 1. The CNR of the T1 weighted image and the SNR of b0 were equivalent for all animals (mean/std=0.7/0.1 and 4.1/0.3 respectively). No correlation was found between age/weight and quality metrics (R<0.55), suggesting that the image quality is not driven by the subject's age or weight in this dataset.
DTI metrics are available for all subjects (Fig.2) along with the averaged template of the weighted images and the computed metrics (Fig.3). We could observe high variance of the data (Fig.3) in the basal ganglia, which can be explained by iron accumulation across age[9]. This variance was mainly observed in the T2 weighted and b0 images, minimally affecting the template of DTI metrics.
The extracted bundles followed the known macaque monkey neuroanatomy (Fig.4): The color-coded concatenation of these callosal streamlines, displayed in the bottom row, corresponds to the frontal, parietal, occipital, and temporal topological organization of the homotopic callosal fibers as previously described[10]. Interested users will be able to replicate the acquisitions on other macaque monkeys and further enrich the MADI@700 dataset. Moreover, the current dataset, already processed and curated for tractography, may be used to investigate microstructural biomarkers.
Nadège CORBIN (TALENCE), Alex VALCOURT CARON, Adrien BOISSENIN, Ankur GUPTA, Tho-Hai NGUYEN, Serge ANANDRA, Sylvain MIRAUX, Maxime DESCOTEAUX, Laurent PETIT, Fabien WAGNER
13:30 - 15:00
#47745 - PG264 A Probabilistic MRI Atlas of the Pituitary Gland in Young Adult Females.
PG264 A Probabilistic MRI Atlas of the Pituitary Gland in Young Adult Females.
The pituitary gland (PG) is a critical endocrine component. It comprises the anterior and posterior lobes (with the less distinct pars intermedia) that serve distinct functions. The anterior PG regulates several neuroendocrine axes, including the hypothalamic-pituitary-gonadal, hypothalamic-pituitary-adrenal, among others. The posterior PG is dedicated to storing and releasing hormones. The PG's size and structure are subject to various influences such as puberty, nutritional status, medication use, and psychiatric disorders, highlighting the importance of detailed volumetric studies. While most studies focus on the entire PG volume, the functional and structural differences between the anterior and posterior lobes suggest that separate analyses could yield more precise insights. Quantitative analysis of brain MRI relies on accurate anatomical segmentation, traditionally achieved through labour-intensive manual labelling. Automated methods offer a more efficient alternative. However, the absence of a standardized PG atlas impedes research in this area. This study aimed to develop optimized spatial normalization protocols for the anterior and posterior PG in healthy young adult females, establishing a crucial reference for future research, particularly in psychiatric conditions prevalent in women, such as anorexia nervosa.
26 healthy young female volunteers (mean age 23±3 years) with normal body mass index (18.5-25.0 kg/m2) were included. Participants had no history of brain or psychiatric disorders and were not using any medications or drugs. High-resolution T1-weighted MRI scans were acquired using a 1.5 T Siemens Sonata system. Manual delineation of the anterior and posterior PG lobes was performed using FSLeyes software. Given the challenges in normalizing the PG within a standard stereotactic space, a three-step optimized spatial normalization procedure was employed. The ICBM152 template in SPM12 was used in the first step, involving linear and non-linear transformations with specific parameters (7×8×7 discrete cosine transform basis functions and 12 iterations). Deformation fields were applied to normalize the binary pituitary delineations, resulting in an initial non-optimized pituitary atlas. A second normalization step, limited to the PG mask, was performed to refine the alignment, addressing the limitations of standard brain masks that exclude the PG. This approach, masking the cost function, optimized the normalization specifically for the PG. A third normalization step further improved the atlas. Atlases for the anterior and posterior PG were created using the spatial transformations from the third normalization step. One participant presented with an intrasellar arachnoidocele, a common condition, and was included in the analysis to reflect population variance. The mean total pituitary volume was 705±88 mm3, with anterior and posterior volumes of 614±82 mm3 and 91±20 mm3, respectively. The probabilistic atlases showed smooth contours and clear boundaries between the anterior and posterior lobes. The global pituitary atlas showed an 80% overlap with manual delineations for the DICE index and 67% for the Jaccard index. For the anterior pituitary atlas, these values were 77% and 64%, and for the posterior PG atlas, 62% and 47%, respectively. Manual delineations closely aligned with the atlas-based estimation of 688±78 mm3, with a non-significant mean relative volume difference of 2.4%. A significant correlation (R=0.44, P=0.025) and good agreement were observed between the two methods. Anterior PG volumes were also similar between manual (615±82 mm3) and atlas-based (607±73 mm3) measurements, with a 1.3% mean relative volume difference and a significant correlation (R=0.43, P=0.027). Although posterior PG volumes were similar between methods (manual: 91±20 mm3, atlas: 92±10 mm3, 1.1% relative difference), the correlation was not significant (R=0.09). There were no significant differences in average anterior-posterior (AP) ratios between manual and atlas-based methods (5.6% mean relative difference), but a strong correlation was observed (ρ=0.54, P=0.004). This study successfully developed the first maximum probability atlas of the pituitary gland, specifically tailored to young adult females. This atlas is a valuable tool for research, particularly in psychiatric disorders with a high prevalence in women, such as anorexia nervosa. The ability to differentiate between the anterior and posterior PG enhances the precision and robustness of research outcomes. The measured PG volumes are consistent with the range reported in other studies, with the reduced variance potentially attributed to methodological improvements or the focus on a homogeneous young female group. The atlas demonstrates good agreement with manual delineations and shows substantial concordance for the entire PG and anterior lobe volumes, as well as AP ratios.
Fabien SCHNEIDER (Saint Etienne), Manel MERABET ZENNADI, Maurice PTITO, Jérôme REDOUTÉ, Nicolas COSTES, Claire BOUTET, Natacha GERMAIN, Galusca BOGDAN
13:30 - 15:00
#47698 - PG265 Open-source simulation tool for characterization of MRI contrast agents.
PG265 Open-source simulation tool for characterization of MRI contrast agents.
Superparamagnetic iron oxide nanoparticles (IONs) have excellent magnetic properties for applications as MRI contrast agents (CA). Relaxometry can improve quantification of ION concentration or spatial distribution, relevant for biomedical research or medical diagnostics. However, the exact relation between changes in relaxation time and CA properties remains elusive [1]. Here, an open-source simulation tool was implemented that enables characterization of relaxation time changes for ION-based CA in MRI.
A Monte Carlo simulation tool was implemented for simulations of the changes in transverse relaxation times T2 and T2* caused by IONs with given properties (Fig. 1) [2]. Inside a cubic voxel (10^-6 – 10^-3 mm^3) IONs were modeled as randomly distributed impermeable spheres, with corresponding dipole fields. Concentration, magnetization, size, and coating of IONs can be defined manually. Diffusion of 10,000 water protons was modeled as random walk for 100 ms. The phase of each water proton spin was accrued over a given number of diffusion steps (10^5 – 10^7) through the local dipole fields. The MR signal at each diffusion step was obtained with an exponential fit of the signal. For T2 simulations, a multi spin echo sequence was implemented, with phase inversions of all spins after each refocusing pulse.
The simulation tool was tested on two ION-based CA, Ferucarbotran (Bayer AG, Leverkusen, Germany) and nanohybrid magnetic nanocubes (MNC@OH) [3]. Ferucarbotran consist of magnetite crystal clusters inside a coating of carboxydextran, with a median hydrodynamic radius of 22.5 nm and magnetization of 380 kA/m (Fig. 2) [4, 5]. MNC@OH were prepared with a ligand exchange procedure using tetramethyl ammonium hydroxide, which results in a median hydrodynamic radius of 177.2 nm and magnetization of 385 kA/m.
For measurements of T2 and T2*, CAs were prepared in agarose phantoms with iron concentrations of 90, 180, 400 µmol/L and 140, 300, 1000 µmol/L for Ferucarbotran and MNC@OH, respectively. Measurements were performed on a 9.4 T animal scanner (Bruker BioSpec 94/20). For T2, a multi spin echo sequence (TE,min = 6.5 ms, echo spacing = 6.5 ms, 16-25 echoes, TR = 2000 ms) and for T2* an ultra short echo sequence (TE,min = 0.45 ms, TE,max = 40 ms, TR = 120 ms, flip angle = 20°) was used.
Simulations of the CA have been performed for three iron concentrations corresponding to the measurements and two approaches for modeling the CA size were tested (Fig. 2C). In the first approach, IONs with uniform radii were simulated, using the median hydrodynamic radius. In a second approach, the distribution of hydrodynamic radii for MNC@OH, and of the magnetite clusters with a coating layer for Ferucarbotran were considered for the simulations. To account for potential particle aggregation of MNC@OH, simulations were also performed with various larger hydrodynamic radii. Measurements and simulations reproduced the linear correlation between concentration and relaxation rates R2 and R2* (Fig. 3). For Ferucarbotran, the simulated R2 and R2*, when using the uniform median radius, were twice as high as the measured values. When the radius distribution and coating were considered, simulation results were in good agreement with the measurements. For MNC@OH, the simulated relaxation rates were similar, irrespective of the approach for modeling the CA size. Deviations between simulated and measured relaxation rates were lowest when the hydrodynamic radius distribution was considered. Simulated R2* fit well to measured values, but for R2 substantial differences were observed. Cryo-electron microscopy (EM) revealed substantial aggregation of MNC@OH (Fig. 4A). Simulations accounting for these larger hydrodynamic radii showed that R2* was independent of the radius, whereas R2 decreased with increasing radius (Fig. 4B). Simulations with Rh = 400 nm reproduced the measured R2* and R2 for MNC@OH (Fig. 4C). The developed simulation tool, with an appropriate approach for modeling CA particle sizes, was able to reproduce experimentally measured R2 and R2* for different ION-based CA. For comparisons of simulated and measured relaxation times, the radius distributions of the CA have to be considered in the simulations. Differences between simulations and measurements of the MNC@OH particles were related to aggregation, confirmed by cryo-EM. The median hydrodynamic radius of these aggregates could be estimated from simulations alone. In this study, an open-source Monte Carlo simulation tool was implemented and validated for simulations of ION-based CA in MRI. This tool can be used to characterize the influence of changes in specific parameters of CA on relaxation time, which was confirmed here by revealing particle aggregation of MNC@OH. Comparison of measured and simulated relaxation times of CA with distinct properties may enable exact quantification of CA concentration and spatial distribution in complex environments, such as tissues of interest.
Lauritz KLÜNDER (Münster, Germany), Bastian MAUS, Maria Belen RIVAS AIELLO, Christian STRASSERT, Cornelius FABER
13:30 - 15:00
#48021 - PG266 Preclinical ORYX-MRS: A toolbox for proton magnetic resonance spectroscopy data analysis in rat brain.
PG266 Preclinical ORYX-MRS: A toolbox for proton magnetic resonance spectroscopy data analysis in rat brain.
Proton magnetic resonance spectroscopy (¹H-MRS) offers non-invasive imaging of brain metabolism [1]. In preclinical neuroscience, rodent models are widely used for understanding disease mechanisms and progression, treatment planning, and studying aging-related diseases [2,3]. Although advanced tools for MR spectroscopy analysis and visualization have been developed for clinical MRS data [4], their applicability to preclinical research remains limited due to differences in data format, anatomical scale, and acquisition protocols. ORYX-MRSI, originally developed for clinical ¹H-MRSI data, integrates features such as spectral visualization, co-registration, and atlas-guided statistical evaluation [5], but requires adaptation for use in rodent studies. The proposed toolbox, Preclinical Oryx-MRS, offers a user-friendly graphical interface and supports key functions such as spectral loading and visualization, co-registration, image registration, segmentation, atlas-based quantification, and metabolite concentration visualization, specifically tailored for rat brain ¹H-MRS data.
Preclinical ORYX-MRS was programmed in MATLAB 2024a (The MathWorks Inc., Natick, MA). The software currently accepts various user-provided input files, including a raw ¹H-MRS data file in .mrd format, an LCModel output .coord file, and a reference anatomical MRI. The toolbox currently includes five main modules: Load Data, Co-registration, Registration, Segmentation, and CRLB & FWHM & SNR.
Load Data:
This module reads and visualizes both raw ¹H-MRS .mrd files and LCModel .coord files. Users can toggle between raw and processed spectral visualizations.
Co-registration:
This module creates a volume of interest (VOI) mask using offset, angle, and size information from the .mrd file. The generated mask is then co-registered to the anatomical reference MRI in NIfTI format. The final mask file is saved as a NIfTI image, and SPM [6] is used to visualize the reference image overlaid with the VOI mask.
Registration:
This module registers the anatomical reference MRI to the SIGMA rat brain atlas [7] using FMRIB's Linear Image Registration Tool (FSL FLIRT) [8]. The resulting transformation matrix is then applied to the VOI mask. Both the registered anatomical image and the mask are then saved in NIfTI format.
Segmentation:
This module uses the anatomical reference image and the SIGMA rat brain atlas segmentation masks to segment white matter (WM), grey matter (GM), and cerebrospinal fluid (CSF) using the SPM toolbox. It then calculates the regional volume fractions and interactively visualizes the results. The segmentation module adapts modified functions from Osprey [9], which was originally developed for clinical MRS data.
CRLB & FWHM & SNR:
This module extracts FWHM and SNR values from the LCModel .table file and plots the CRLB and concentration values for selected metabolites.
Toolbox Assessment:
A rat brain was scanned on a 7T preclinical MR scanner (MR Solutions, Guildford, UK). T2-weighted anatomical MRI (TR/TE = 5000/45 ms, slice thickness = 1 mm) and 1H-MRS data (PRESS (Point Resolved Spectroscopy), 3x3x4 mm3 voxel, TR/TE= 3000/24 ms, 2048 time points, 256 averages) were acquired. The .mrd formatted MRS file, .nifti formatted T2w MRI, and a .coord file from an open source [10], were used to evaluate the toolbox. Figure 1 shows the interface of the toolbox, highlighting five of its modules and shows visualizations of a raw ¹H-MRS .mrd file and an LCModel .coord file. Figure 2 displays the VOI mask co-registered to the T2-weighted rat brain MRI, which was then registered to the SIGMA atlas. Figure 3 illustrates the segmented WM, GM, and CSF, their corresponding volume fractions, and the FWHM & SNR plots. Preclinical ORYX-MRS is a user-friendly ¹H-MRS data analysis toolbox developed as an extension to the ORYX-MRSI platform. It currently features five modules for reading and visualizing MR spectral data, creating and co-registering VOI masks, registering anatomical MRI and masks to the SIGMA atlas, segmenting WM, GM, and CSF, calculating tissue fractions, and plotting LCModel-derived quality metrics. Future updates will include LCModel-based quantification of .mrd files and a region-based data analysis module, and data distribution plots. Preclinical ORYX-MRS provides a solution for the analysis and visualization of preclinical ¹H-MRS data in rat brain studies. By integrating essential functions such as spectral visualization, VOI mask generation and registration, tissue segmentation, and LCModel-based quality metric extraction, the toolbox will aid preclinical MRS workflows.
Melisa ÖZAKÇAKAYA (Istanbul, Turkey), Sevim CENGIZ, Arnold BENJAMIN, Uluç PAMUK, Oğuzhan HÜRAYDIN, Pınar ÖZBAY, Ruslan GARIPOV, Esin OZTURK ISIK
13:30 - 15:00
#47812 - PG268 A real-world patient MR Angiography (MRA) Database for Brain Vasculature Segmentation and Modelling in Predictive Simulation for the Planning of Interventional Neuroradiology procedures (PreSPIN).
PG268 A real-world patient MR Angiography (MRA) Database for Brain Vasculature Segmentation and Modelling in Predictive Simulation for the Planning of Interventional Neuroradiology procedures (PreSPIN).
Ischemic stroke (IS) is a major cause of disability and death worldwide. Endovascular Thrombectomy(EVT) has proven to be highly effective, and is now a standard clinical intervention [1]. Baseline(D0) and 24hrs follow-up(D1) MR or CT data are routinely acquired. MR angiography (MRA) is often included in the routine protocol, and sometimes with MR perfusion to confirm diagnosis or treatment decision [2].
A safe and efficient EVT requires a strict management timeline and highly skilled interventionalists. Training is a key element of performance and process improvement due to the need of complex microcathether navigation into the intracranial circulation. Recent international recommendations(WFITN) integrated simulation in the training procedure [3]. The planning phase of EVT may also benefit from simulation with predictive capabilities, e.g. to plan catheter navigation (mechanical simulation of the interventional devices), to improve perfusion imaging or to predict reperfusion (computation of fluid dynamics). New brain vasculature segmentation and modelling methods are therefore necessary to provide geometric boundary conditions for numerical predictive simulation, and the dedicated real-world patient image database is necessary.
While several brain vasculature databases exist [4], real-world brain MRA database from acute ischemic stroke phase is missing. Clinical images are often difficult to obtain. In this work, we present how we constructed such a MR angiography database, named PreSPIN (Predictive Simulation for the Planning of Interventional Neuroradiology procedures), from real-world clinical images. Vessel segmentation obtained from fine-tuned deep learning segmentation algorithm is also included.
Database specifications: The database construction is based on the following inclusion criteria to balance between the clinical scenario and algorithm development requirement: 1).MR field strength used in everyday practice; 2).MR 3D Time of Flight(TOF) images with diagnostic quality; 3).Patients with both D0 and D1 TOF availability; 4).D0 MR perfusion data availability. Thanks to our research image data platform ArchiMed[5], EVT related multi-centric clinical trials are archived together including metadata, and candidate patients for this database can be easily identified. Quality control was conducted and saved as metadata.
Database construction procedure: Selected DICOM headers (magnetic field strength, image resolution, and brain coverage) of all IS patient MR TOF data were analysed to characterise in the year (2021) before building the database (2022).Only exams with a diagnostic image quality noted at least as “good” were selected. They were then sorted according to: D0 and D1 availability, then D0 availability; decreasing voxel isotropic ratio (VIO, defined in Figure 1 c). Exams with insufficient 3D TOF coverage were also excluded. This was assessed using MIP visualization in 3D Slicer, with a preference for full head coverage, and at minimum full visibility of the Circle of Willis and major arteries of the anterior and posterior circulation. 3D TOF data from EVT patients did not cover the full head and was considered non-identifiable. The data were anonymised and saved in BIDS format. They were also detached from clinical trials. A pretrained deep learning (DL) algorithm [6, 7] was further fine-tuned and applied to this database for future expert modification. A total of 210 MR 3D TOF data were identified (acquired between 09/2021 and 03/2023), involving 161 patients (mean age: 65yrs; 104 male, 55 female), with a subset of 49 patients (mean age 63yrs, 32 males vs 17 females) who had both D0 and D1 data. Data acquisition was performed on 1.5T and 3T systems in a ratio of 65% to 35%, consistent with our investigation for data in 2021(68% 1.5T, 32% 3T). Additionally, 54 D0 MR perfusion datasets were identified, among which 26 patients had corresponding D0 data included. Initial segmentation of all TOF data were incorporated into the database. Database characteristic histograms are shown in Figure with the age, image resolution and VIR distribution. Real-world patient image data are important for algorithm development. We have shown that our database reflected clinical practice. Future updates will include expert corrections to the initial DL algorithm output segmentation.Indeed, any segmentation is error prone, and what is considered a good segmentation might depend on the targeted clinical application [8]. The PreSPIN database has the potential to serve as a collaborative space where multiple experts can provide their correction to the same exam, fostering future algorithm development. The PreSPIN MRA database is well-structured and close to real-world clinical imaging data. It can serve as a benchmark for testing different brain vascular segmentation and modelling algorithms.
Bailiang CHEN (Nancy), Odyssée MERVEILLE, Emilien MICARD, François ZHU, Ulysse PUEL, Pierre ROUGE, Nicolas PASSAT, Benjamin GORY, Erwan KERRIEN, Marine BEAUMONT
13:30 - 15:00
#47803 - PG269 PatientSpace : an interpretable graph-based latent space for multimodal neuroimaging biomarker learning in Alzheimer’s Disease and Frontotemporal dementia.
PG269 PatientSpace : an interpretable graph-based latent space for multimodal neuroimaging biomarker learning in Alzheimer’s Disease and Frontotemporal dementia.
Alzheimer’s disease (AD) and frontotemporal dementia (FTD) are complex and heterogeneous neurodegenerative disorders, each characterized by variability in clinical presentation, genetic factors, and neuroimaging profiles [1], [2], [3], [4]. The overlap in symptoms and imaging features between AD and FTD complicates the diagnosis [5]. To address this challenge, we propose PatientSpace, a graph-based representation of a structured and interpretable latent space built from multimodal neuroimaging data, aimed at capturing disease heterogeneity and facilitating subtype discovery across both AD and FTD.
We analyzed 1,522 18F-FDG PET and MRI scans, split into training (60%), validation (20%) and test (20%) sets, stratified by diagnosis and age to avoid subject overlap. The model was trained using data from 336 cognitively normal (CN), 504 AD and 106 FTD subjects. We extended the DIVA framework [6] to a multimodal setting, integrating auxiliary tasks for diagnosis and age prediction, along with latent space consistency constraints. A graph was constructed from the latent space using a dissimilarity measure derived from latent consistency. PatientSpace was evaluated through (1) agglomerative clustering over the graph and (2) local neighborhood analysis on an unseen test set. Additionally, longitudinal scans from 673 mild cognitive impairment (MCI) subjects were projected into the latent space to study dementia conversion. PatientSpace effectively distinguished CN, AD and FTD groups, achieving classification performance comparable to state-of-the-art approaches : CN (Sensitivity: 0.99 - Recall: 0.87); AD (Sensitivity : 0.91; Recall : 0.92) and FTD (Sensitivity 0.70; Recall : 0.91). Ten distinct clusters were identified, differing in terms of diagnosis, age, MMSE, atrophy and metabolism patterns. Five AD clusters have been identified. All exhibited typical patterns of brain atrophy and metabolic changes, except for two clusters – one showing only atrophy without distinct metabolic pattern, and the other lacking any clear pattern when compared to CN groups. Additionally, two FTD clusters emerged. One aligned with the classical FTD profile, displaying frontal atrophy and hypometabolism, while the other presents a less typical pattern, characterized by temporal atrophy and hypometabolism without involvement of the frontal regions. Neighborhood analysis demonstrated coherent spatial organization with similar subjects sharing neuroimaging features. MCI projections revealed distinct progression trajectories, with most subjects initially positioned closer to CN clusters, and early separation between converters and non-converters observable at baseline. PatientSpace provided a unified representation of neurodegenerative diseases, capturing both global and local heterogeneity. While the framework successfully differentiates AD, FTD and CN populations, limitations include class imbalance, potentially affecting FTD classification performance, and modality-related sensitivity differences, as PET may detect earlier changes than MRI [7]. Discrepancies with previously reported AD subtypes may stem from differences in input modalities, model strategies and cohort composition [8], [9]. MCI projections supported the model’s potential to detect early conversion risk through multimodal embedding. We present PatientSpace, a graph-based latent space framework that enables interpretable, multimodal representation learning for the study of AD and FTD. By capturing subject-level heterogeneity and disease trajectories, PatientSpace offers new opportunities for data-driven subtyping and personalized analysis of neurodegeneration.
Dorian MANOUVRIEZ (LILLE), Grégory KUCHSINSKI, Simon LECERF, Hélène LAHOUSSE, Maxime BERTOUX, Antoine ROGEAU, Thibaud LEBOUVIER, Vincent ROCA, Renaud LOPES
|
Poster hall |
14:00 |
"Thursday 09 October"
Added to your list of favorites
Deleted from your list of favorites
E12
14:00 - 15:00
MS5 - Magnetic Resonance Imaging Biomarkers
For early treatment response in radiotherapy
|
Salle 76 |
15:00 |
TIME FOR A BREAK
- Coffee and refreshments will be available at the cash bar.
|
15:30 |
"Thursday 09 October"
Added to your list of favorites
Deleted from your list of favorites
A16
15:30 - 17:00
FT3 ORAL - Reliably probing the neural tissue
FT3: Cycle of Quality
15:30 - 15:40
#45941 - PG007 A systematic review of cerebrovascular reactivity delay assessment in gas-challenge blood-oxygen-level-dependent magnetic resonance imaging.
PG007 A systematic review of cerebrovascular reactivity delay assessment in gas-challenge blood-oxygen-level-dependent magnetic resonance imaging.
Cerebrovascular reactivity (CVR), the ability of cerebral blood vessels to respond to vasoactive stimuli, is a key marker of vascular health [1]. One commonly utilised method for measuring CVR is blood-oxygen-level-dependent magnetic resonance imaging (BOLD-MRI) during vasoactive gas administration [2]. CVR magnitude, which assesses the vascular response size in relation to the stimulus, is a well-established metric associated with various pathologies [1]. However, temporal aspects, such as CVR delay—the interval between stimulus onset and vessel response—are comparatively underexplored [1]. This lack of research is partly due to inconsistencies in methodologies and definitions, which hinder the use of delay as a neurovascular integrity metric. To address this issue, we conducted a systematic review to identify and synthesise studies that explore temporal CVR features, summarising key findings and highlighting gaps in the current literature.
We systematically searched the EMBASE, MEDLINE, Web of Science, and the Institute of Electrical and Electronics Engineers (IEEE) Xplore databases for publications from January 1990 to April 2025. Our search strategy targeted human CVR studies using gas-challenge BOLD-MRI that explicitly addressed temporal features of the BOLD response. Studies that corrected for CVR delay in their methods, even without using delay as an outcome variable, were included. Deduplication was carried out using the ASySD software [3] alongside manual inspection. One author (K.N.R.) performed title/abstract and full-text screening, with any eligibility queries resolved through discussions with other authors. We extracted data encompassing demographics, CVR tolerability, MRI parameters, paradigm features, delay computation methods, reported delay values, and delay-relevant findings. We identified 190 eligible studies comprising 5575 unique participants (3806 patients and 1769 healthy individuals) and 6149 scans. The average sample size per study was 46 participants, with a mean age of 46.0 years and a sex distribution of 54.2% males and 45.7% females. Most studies utilised 3T MRI alongside a hypercapnic gas challenge, though the gas delivery methods varied. Both end-tidal targeting and fixed-composition gas administration were widely employed (Figure 1). Paradigm design and duration also varied among studies; block-type paradigms typically lasting 5-10 minutes were the most popular (Figure 2). Several delay computation techniques were reported, including cross-correlation [4], iterative delay-to-plateau (DTP) determination [5], linear regression [2], and haemodynamic response function (HRF) fitting [6] (Figure 3). However, descriptions of these delay methods often lacked detail, and studies comparing different delay computation methods were scarce (6 studies, 3%). These studies analysed numerous techniques and generally found that voxel-wise and model-based approaches outperform global methods. Only four (2%) studies evaluated delay validity by assessing repeatability or comparing similar measures from different MRI sequences. Out of 137 studies focused on pathology, only 34 (25%) used CVR delay as a variable of interest. In these studies, patients with steno-occlusive disease, sickle cell disease, and Alzheimer's disease typically exhibited longer delays compared to healthy controls, while results regarding delays in other conditions were more mixed (Table 1). Differences in experimental and processing methods, as well as patient demographics, were prevalent across studies examining the same pathology. The findings of our review highlight the significant variety in techniques for computing CVR delay using gas-challenge BOLD-MRI data. We found considerable heterogeneity in gas administration methods and paradigm designs. Although longer CVR delays are reported in certain diseases, inadequate reporting and the lack of methodological standardisation limit the ability to draw conclusions regarding their clinical importance. Future studies should focus on establishing, comparing, and validating standardised methods for evaluating delay and other temporal CVR characteristics, as well as investigating the influence of CVR experimental factors on delay values. Delay application in large-scale studies is needed to establish CVR delay ranges in healthy and patient populations and to thoroughly assess the utility of CVR delay as a clinical biomarker. Our systematic review emphasises the need for harmonised approaches to calculate CVR delay using gas-challenge BOLD-MRI data. Although some evidence indicates longer delays in select pathologies, current methodological inconsistencies have hindered the wider application of CVR delay in clinical research. Standardising and validating methodologies across a broad range of diseases is essential to establish CVR delay and other temporal CVR characteristics as robust biomarkers for cerebrovascular health.
Keelin RIDGE (Edinburgh, United Kingdom), Joanna WARDLAW, Michael THRIPPLETON, Michael STRINGER
15:40 - 15:50
#47859 - PG008 Multi-sequence, multi-center arterial spin labelling-based cerebrovascular age prediction and normative perfusion models.
PG008 Multi-sequence, multi-center arterial spin labelling-based cerebrovascular age prediction and normative perfusion models.
Cerebral blood flow (CBF) declines in healthy ageing2 and is altered in many brain pathologies1. Thus, pathological CBF deviations should be normalised against age-related CBF change, for which two methods currently exist. Brain-predicted age gap (BAG) estimation methods identify accelerated brain ageing compared to a healthy population by using neuroimaging-derived structural features and machine learning, and higher than chronological brain ages have been associated with cognitive decline and mortality3,4. BAG estimation methods have recently included arterial spin labelling (ASL)-derived features such as CBF, dubbed ‘cerebrovascular age’, and showed improved estimation accuracy5 and differentiation between healthy controls and Alzheimer’s disease6. These studies used single-sequence datasets and utilising larger harmonised cohorts may improve estimation accuracy. Normative modeling is a framework that maps healthy perfusion change across age, at which differences at the level of a single subject can be determined7, which has not yet been applied to ASL-derived CBF. Both cerebrovascular age, performed in a single center, and normative models could improve with larger datasets, as single-centre CBF studies are limited in their applicability to other cohorts due to MRI sequence differences, which affect perfusion quantification8.
We will investigate the possibility of multi-center cerebrovascular brain age modeling and normative modeling, as well as assess the benefit of harmonization.
Five cohorts (n=2925) differing in age and sex distributions (Table 1), and 3T pseudo-continuous ASL sequences (3D FSE spiral, 2D EPI, 3D GraSE, Table 1) along with T1-weighted and FLAIR scans were included. Image processing was performed using ExploreASL9, providing single-compartment quantified regional CBF and spatial coefficient of variation (sCoV) values from the total grey matter (GM) and vascular territories.
Next, ExtraTrees-derived BAGs were obtained by training in the training cohort, and testing in EDIS, HELIUS, Insight 46, and EDIS cohorts using the CBF and sCoV features for the unharmonised and harmonised datasets separately. Normative models were created used generalised additive models (GAM) of GM CBF.
CBF and sCoV values were harmonised using several methods, including NeuroComBat, CovBat, NeuroHarmonize, OPNested ComBat, AutoComBat, and RELIEF10. While most methods utilise a single batch parameter, both OPNested ComBat and AutoComBat allow for multiple batch parameters for which labelling duration, post-labelling delay and readout type were used, and RELIEF utilises a latent-space assessment of batch effects.11. For the normative model, GM CBF was harmonised with NeuroComBat and through Location, Scale and Shape (LSS) adjustments common in GAM models.
Harmonisation methods were statistically compared for reducing cohort differences in GM CBF, as well as BAGs (predicted minus chronological age) and their mean absolute error (accuracy of the model, MAE) across methods. The normative models were visually evaluated for differences in their associations between age and GM CBF. GM CBF differed between 9 out of 10 cohort-pairs (p<0.001) before harmonisation, and between 6-7 cohort-pairs (p < 0.001) except for RELIEF (no cohort-pairs, p > 0.99) after harmonisation (Figure 1).
The BAGs decreased from 0.6±13.4 years to -1.5±7.9 years for NeuroComBat, around -1.9±7.8 years for CovBat, NeuroHarmonize, and OPNested ComBat, -1.0±7.9 years for AutoComBat, and -5.1±9.4 years for RELIEF (Figure 2). The MAE improved from 11.1±7.5 years to around 6.4±4.9 years for NeuroComBat, CovBat, NeuroHarmonize, OPNested ComBat, and AutoCombat, and 8.8±6.2 years for RELIEF (Figure 2).
The normative models demonstrated a non-linear relationship between GM CBF and age after harmonisation using NeuroComBat or LSS, while the unharmonised model showed a linear relationship (Figure 3). The female models showed a higher CBF compared to males. Harmonisation using ASL sequence parameters improved harmonisation nominally more than conventional methods, while latent-factor approaches appear less accurate, suggesting single batch descriptions do not fully capture differences in perfusion distributions due to ASL sequence parameters, which in turn affects cerebrovascular age estimations. EDIS showed most change with harmonisation, possibly due to the short PLD in EDIS. The training cohort appears to be negatively affected in its CBF and BAG distribution, possibly through transfer of ‘less healthy’ distribution characteristics to the training dataset. The normative models show a non-linear association between age and GM CBF, conflicting with cross-sectional studies2, while no association between age and CBF at older age was also found12, partly explaining the apparent increasing slope of CBF decline in our findings. Multi-sequence, multi-cohort ASL-derived cerebrovascular brain age and normative models are feasible using ASL-parameter specific harmonisation.
Mathijs DIJSSELHOF (Amsterdam, The Netherlands), Candace MOORE, Saba AMIRI, Beatriz PADRELA, Mervin TEE, Saima HILAL, Christopher CHEN, Bert-Jan H. VAN DEN BORN, Wibeke NORDHØY, Ole A. ANDREASSEN, Lars T. WESTLYE, Nishi CHATURVEDI, Alun D. HUGHES, David M. CASH, Jonathan M. SCHOTT, Carole H. SUDRE, Frederik BARKHOF, Joost P.a. KUIJER, Francesca BIONDO, James H. COLE, Henk J.m.m. MUTSAERTS, Jan PETR
15:50 - 16:00
#47875 - PG009 Feasibility of single-bolus MR Vascular Fingerprinting with spin-and-gradient-echo DSC-MRI.
PG009 Feasibility of single-bolus MR Vascular Fingerprinting with spin-and-gradient-echo DSC-MRI.
Optimal treatment planning for patients with adult-type diffuse glioma remains hindered by the need for invasive tissue analysis. Non-invasive glioma diagnosis can potentially be achieved by probing the tumor vasculature using advanced Dynamic Susceptibility Contrast MRI (DSC-MRI) [1–3] with MR vascular fingerprinting (MRVF) [4,5]. As reduction of contrast agent usage gains traction in the clinic [6], investigating the feasibility of single-bolus MRVF is an essential step towards clinical translation.
In a recent simulation study, we found that omission of the preload bolus seems feasible, albeit with a penalty on the accuracy for determining the rCBV [7]. Currently, we aim to do an in vivo comparison of the parameter maps estimated with a single bolus and a double bolus.
This prospective study was performed as part of the IRB-approved Vascular Signature Mapping (VSM) study (ClinicalTrials.gov ID: NCT05274919), and written informed consent was obtained from all patients. Scanning was performed at 3T (GE, Milwaukee, WI, USA) in eight patients under surveillance after previous glioma treatment (5 female, mean age 47 years). The protocol included two spin-and-gradient-echo (SAGE) DSC scans during each of which a bolus of 7.5 mL Gadovist was injected. Imaging parameters were: TR = 1500 ms, TE(GRE)/TE(SE) = 17/70 ms, voxel size = 2.3 x 2.3 x 3.0 mm3, with 120 time points and 15 slices covering the tumor area or resection cavity. The MRVF dictionary was obtained using a toolbox [8], simulating the spin- and gradient echo signal for varying rCBV (0.5%–10%, 40 logarithmically-spaced values), vessel radius (3.8-150 μm, 54 logarithmically-spaced values) and permeability (0–0.0060 s−1, 11 values). The matching was performed by minimizing the squared Euclidean distance between the baseline-normalized dictionary atoms and the measured signal time courses. Furthermore, vessel size imaging (VSI) [2,9] maps were obtained using in-house code developed by Arzanforoosh et al. [1]. These vessel size maps were visually compared to the MRVF maps with a single bolus (i.e. from the first SAGE-DSC scan) and a double bolus (i.e. second SAGE-DSC scan). The anatomical scans and MRVF parameter maps for a patient with contrast enhancement are shown in Figure 1. Visually, the rCBV and vessel radius maps with a single bolus and a double bolus bolus exhibit a large similarity, while the estimated permeability values are notably higher in case of the double bolus. Furthermore, the regions of contrast enhancement on the T1w scan are reflected in the permeability maps.
Figure 2 shows the distribution of values for the three parameters in normal-appearing gray matter across all patients, comparing the single- and double- bolus case. The spread and the median value for the vessel radius increases slightly in case of a double bolus. Permeability estimates are mostly zero for the single bolus, while there is substantial spread in case of a double bolus. For the rCBV estimates, the distribution appears to remain similar.
Figure 3 shows the vessel size maps as obtained using MRVF and VSI with a single bolus and double bolus strategy, for the same patient as Figure 1. It can be observed that estimations between the single and double dose vary only slightly. Furthermore, VSI tends to estimate larger vessels than MRVF, and larger vessels tend to be estimated in the gray matter compared to the white matter. While both permeability maps in Figure 1 reflect the contrast-enhancing regions of the T1w scan, the posterior lesions with lower contrast enhancement had improved visibility in case of a single bolus. Furthermore, as the permeability is expected to be zero outside of the (resected) tumor area due to an intact blood-brain-barrier, we consider the map with a single bolus to be a more truthful estimation. In contrast, the estimations of vessel radius and rCBV gave comparable maps with the single and double bolus.
Further investigation is needed in more patients - preferably including preoperative patients - to validate the findings. Also, a statistical analysis and a comparison of the rCBV and permeability maps to reference techniques must be performed. MRVF is a promising technique to quantify the vascular architecture in the brain. Our preliminary results indicate that estimation of the vascular maps with a single bolus of contrast agent can lead to similar results for rCBV and vessel radius. For the permeability maps, the substantial differences seem to point at an improved estimation for a single bolus.
Karen VAN DER WERFF (Rotterdam, The Netherlands), Danielle VAN DORTH, Krishnapriya VENUGOPAL, Marion SMITS, Stefan KLEIN, Esther WARNERT, Juan HERNANDEZ-TAMAMES, Matthias VAN OSCH, Frans VOS, Dirk POOT
16:00 - 16:10
#46912 - PG010 Feasibility and Hemodynamic Insights from Single- and Multi-Delay ASL MRI in Preterm Neonates.
PG010 Feasibility and Hemodynamic Insights from Single- and Multi-Delay ASL MRI in Preterm Neonates.
Preterm neonates are particularly vulnerable to brain injury linked to cerebral blood flow (CBF) disruption. Arterial Spin Labeling (ASL) MRI offers a non-invasive and quantitative method for assessing cerebral perfusion but remains underutilized in neonatology due to concerns about motion artifacts and low signal-to-noise ratio (SNR). In this study, we evaluate the feasibility and performance of single- and multi-delay pseudo-continuous ASL (PCASL) MRI in non-sedated preterm neonates, comparing scan quality and hemodynamic values across acquisition types and examining associations with age, sex, and fetal-type variants.
Forty-eight preterm neonates (25 male, mean gestational age 28.7±2.6 weeks, postnatal age 9.7±5.0 weeks) underwent structural MRI (T1w, T2w) and three ASL protocols: single-delay (PCASL_1PLD), three-delay (PCASL_3PLD), and seven-delay (PCASL_7PLD). Scan quality was independently rated (“good,” “acceptable,” “unusable”) and compared across modalities. Cortical CBF and arterial transit time (ATT) were quantified and compared using t-tests and Cohen’s d across modalities. Hemodynamic parameters were further correlated with postmenstrual age (PMA), postnatal age (PNA), gestational age (GA), sex, and fetal-type posterior cerebral artery variants. Image Quality:
PCASL_3PLD yielded the highest quality, with <10% unusable scans and >90% rated “good” or “acceptable.” PCASL_1PLD was most affected by motion, while PCASL_7PLD had cases of low SNR. T2w scans outperformed T1w scans (only 4.2% vs. 25% unusable, p<0.01). When structural scans were rated unusable, ASL scans remained interpretable in over half of cases (50% of PCASL_1PLD, 41.6% of PCASL_3PLD, and 66.6% of PCASL_7PLD scans were of “good” or “acceptable” quality), highlighting ASL's complementary value.
CBF Measurements:
PCASL_1PLD underestimated cortical CBF compared to multi-delay PCASL (CBF_1PLD: 13.1±4.3 vs. CBF_3PLD: 18.7±5.6 and CBF_7PLD: 20.6±6.1 ml/100g/min, p<0.001, d≥1.12). CBF_1PLD was significantly lower in females (p=0.035), but no sex differences were observed in CBF_3PLD or CBF_7PLD. CBF asymmetry across hemispheres was not significant. Bland-Altman analysis showed the greatest discrepancy between CBF_1PLD and CBF_7PLD (mean bias 8.1 ml/100g/min). CBF_1PLD and CBF_3PLD were significantly correlated with PNA and PMA. No significant predictors were found for CBF_7PLD.
ATT Measurements:
ATT_3PLD and ATT_7PLD were similar (1298 ms vs. 1315 ms, p=0.77). ATT was longer in males (p≈0.05, d=0.60), suggesting sex-dependent differences in blood arrival time. No significant associations were found between ATT and age or birth indication. Our findings demonstrate the feasibility and diagnostic potential of multi-delay PCASL, particularly PCASL_3PLD, in non-sedated preterm neonates. Compared to single-delay PCASL, multi-delay protocols offered higher image quality, more reliable CBF estimates, and greater resilience to motion artifacts. PCASL_3PLD achieved an optimal balance of scan duration and SNR, outperforming PCASL_1PLD, which was prone to motion artifacts, and PCASL_7PLD, which suffered from low signal robustness. The CBF underestimation in single-delay ASL is likely attributable to prolonged ATT in neonates, highlighting the limitations of shorter PLD protocols in this population.
Sex-related differences in ATT and CBF_1PLD — but not in multi-delay CBF — suggest that quantification bias may confound interpretations from PCASL_1PLD, reinforcing the importance of delay-sensitive acquisitions. The observed associations between CBF and both PNA and PMA support the developmental sensitivity of perfusion imaging. Despite some variability in signal, PCASL scans were interpretable even when structural scans failed, making it a promising modality for fragile neonatal populations. Multi-delay ASL, particularly PCASL_3PLD, is feasible and advantageous in preterm neonatal brain imaging, offering robust image quality and age- and sex-sensitive hemodynamic assessments. These findings support its integration into neonatal imaging protocols for improved assessment of cerebral perfusion and risk stratification in early neurodevelopment.
Yeva PRYSIAZHNIUK, Yeva PRYSIAZHNIUK (Prague, Czech Republic), Sasha ALEXANDER, Rui Duarte ARMINDO, Elizabeth TONG, Kristen W YEOM, Jakub OTÁHAL, Martin KYNČL, Michael MOSELEY, Gary K STEINBERG, Jan PETR, Moss Y ZHAO
16:10 - 16:20
#47339 - PG011 Fast oxygen metabolism quantification using dual-echo pseudo-continuous arterial spin labeling with a hyperoxic respiratory challenge.
PG011 Fast oxygen metabolism quantification using dual-echo pseudo-continuous arterial spin labeling with a hyperoxic respiratory challenge.
Cerebral blood flow (CBF) and volume (CBV) are highly important cerebral perfusion parameters for multi-parametric quantitative BOLD (mqBOLD) MRI. [1,2] They are commonly mapped by arterial spin labeling (ASL) [3] and contrast-agent-based dynamic susceptibility contrast (DSC) MRI [4], respectively. In mqBOLD, the oxygen extraction fraction (OEF) is calculated from the transverse relaxation rate R2’ and CBV and then combined with CBF to yield the cerebral metabolic rate of oxygen (CMRO2). [1,2,5] We propose to estimate both CBF and CBV non-invasively from a single measurement by combining dual-echo pseudo-continuous ASL (de-pCASL) with a hyperoxic respiratory challenge. Hereby, an ASL-based CBF measurement (first echo, TE1) [3] is combined with hyperoxic BOLD-fMRI (second echo, TE2) for CBV quantification [6]. Hyperoxic de-pCASL not only allows for reducing scan time in mqBOLD by obtaining CBF and CBV from a single scan, instead of two separate measurements, but also eliminates the need for contrast agent application in DSC-based CBV. In this work, we demonstrate the feasibility of de-pCASL with a hyperoxic challenge for calculating CMRO2 from the resulting CBF and CBV estimates using the mqBOLD approach.
13 healthy subjects (29.6±11.4 years) underwent MRI on a clinical 3T system (Ingenia Elition X, Philips, NL) with a 32-channel head coil (Fig. 1). De-pCASL data were acquired with a hyperoxic respiratory challenge (1 min air, 3 min 100% O2) and end-tidal gas sampling (ML206, ADInstruments, NZ). T2- and T2*-weighted data were acquired using DL-accelerated (Philips SmartSpeed, R = 4) [7] multi-echo 3D GRASE and GRE, respectively, for relaxometry.
Data processing and analysis were performed using SPM12 [8], FSL [9], and custom MATLAB programs (R2021b, The MathWorks Inc., USA). De-pCASL echoes were processed separately: TE1-data were evaluated during the normoxic period to calculate CBF according to recommendations [3]. TE2-data were used to estimate CBV using the model by Blockley et al. [6]. T2 and T2* were obtained by exponential fitting of multi-echo relaxometry data. Resulting parameter maps were combined according to Fig. 1 to yield OEF and CMRO2. [1,2] All parameter maps were analyzed in cortical GM (cGM) volumes-of-interest (VOI). Fig. 2 shows subject average parameter maps. Perfusion maps (B/C) show higher values in GM than WM. The R2’ map (A) exhibits strong hyperintensities along the brain’s outline, which propagate into OEF and CMRO2 maps (D/E). Average parameter values in cGM are CBF = 33.1±5.6 ml/100g/min, CBV = 2.1±0.3 %, R2’ = 5.4±0.4 Hz, OEF = 0.90±0.11, and CMRO2 = 583.8±116.4 umol/100g/min (Tab. 1). Average perfusion parameter maps from hyperoxic de-pCASL (Fig. 2B/C) show the expected contrast with noticeably higher perfusion in GM. Regarding CBF, our values agree with literature (Tab. 1A/C) [1,10-13] but lie at the lower end of the spectrum, which may be due to non-optimal background suppression [14,15]. Average CBV is comparable to values from hyperoxic BOLD-fMRI (Tab. 1B) [6,16-18]. Generally, the hyperoxia method yields CBV values of deoxygenated blood, i.e., primarily from the venous compartment, which constitutes about 50-70% (without/with capillary contribution) of total CBV [19]. This explains the deviation from DSC-based estimates, which include contributions from the entire vasculature (Tab. 1C) [1,2,5,13,20].
DL-accelerated relaxometry yields R2’ estimates comparable to literature using similar methods (separate measurements of T2 and T2*), as previously validated [2,5,13,16,17,20]. However, these R2’ values are significantly higher than R2’ estimates obtained from gradient echo sampling of the spin echo (GESSE) or asymmetric spin echo (ASE) [20,21]. Combining R2’ and venous CBV results in unphysiologically high OEF values, which have also been reported previously both in combination with hyperoxia CBV estimates and R2’ from separate T2 and T2* mapping [16,17,20]. This further propagates into CMRO2 values, which clearly surpass literature (Tab. 1C) [1,13]. The hyperintensities observed along the brain’s outline (Fig. 2A/D/E) are mainly due to T2* underestimation caused by strong uncorrected magnetic field inhomogeneities at the brain surface and possible incomplete motion correction. [22,23] Hyperoxic de-pCASL enables simultaneous acquisition of CBF and CBV. Individual parameter values are comparable to separate measurements by (de-)pCASL for CBF and hyperoxic BOLD-fMRI for CBV. However, as reported previously, quantifying advanced oxygen metabolism parameters like OEF and CMRO2 by the mqBOLD technique remains challenging.
Elisa SAKS (Munich, Germany), Stephan KACZMARZ, Nicholas BLOCKLEY, Christine PREIBISCH, Gabriel HOFFMANN
16:20 - 16:30
#46753 - PG012 ON-Harmony: A multi-site, multi-modal travelling-heads resource for brain MRI harmonisation.
PG012 ON-Harmony: A multi-site, multi-modal travelling-heads resource for brain MRI harmonisation.
MRI quantifiability/repeatability is hindered by non-biological sources of variability, e.g. scanner hardware/software [1], [2], [3]. Several approaches exist which attempt to harmonise acquisition and processing [4], [5], including vendor-agnostic sequences [6] and modelling/removing non-biological batch effects [7], [8]. To map these confounding factors for neuroimaging studies, we ran a multi-modal travelling heads study. Healthy participants were scanned at different 3T MRI systems of several types and vendors across different sites. ON-Harmony [9], [10] (Fig1a-b) is one of the most comprehensive travelling-heads resources: 20 subjects, each scanned in up to 8 3T scanners (from 10 considered scanners), 3 vendors (Siemens/Philips/GE) and 5 modalities (T1w-MPRAGE, T2w-FLAIR, diffusion MRI, resting-state fMRI, swMRI). 9 subjects were scanned repeatedly in the same scanner, enabling within-scanner, between-scanner and between-subject variability to be mapped across hundreds of multi-modal imaging-derived phenotypes (IDPs). We have recently scanned 11 of the subjects (1 with repeat scans) at the Stockport UK Biobank (UKB) imaging centre (Reading UKB imaging centre also planned), enabling linkage of UKB population-level imaging data with various clinical scanners. Here we showcase the data and reuse scenarios for assessing the efficacy of harmonisation approaches. ON-Harmony is publicly released [9], [11].
ON-Harmony consists of 2 primary phases (10 subjects each), plus the UKB extension. Phase characteristics and demographics are summarised in Fig1c. Acquisition protocols were aligned with the UKB imaging study [12], while respecting best practices and hardware limitations for scanners (i.e. parameters not simply nominally-matched) [10]. Each subject was scanned in 6 different scanners and 9 subjects had 5 additional within-scanner repeats. 11 participants were additionally re-scanned at the UKB centre, and 1 subject had an addition 5 within-scanner repeats. Other than the UKB data (currently being processed), all data underwent quality control through visual inspection and then using MRIQC [13] (T1w, T2w and fMRI) and eddyQC [14] (dMRI), providing image-quality metrics (IQMs). Data were processed with a modified version [10] of the UKB pipeline [15]. Hundreds of IDPs were derived for each session, allowing us to quantify IQM/IDP variability. Fig2a shows a subject’s raw data, depicting all sessions across scanners (columns) and modalities (rows). IQMs were used to assess consistency of data quality across subjects and scanners (Fig2b). The plots indicate no major outlier scanners/subjects in terms of image quality across phases and modalities.
We assessed IDP similarity for within-scanner, between-scanner, between-subject sessions. Fig3a shows between-session similarity matrices for a subset of participants with within-scanner repeats, showcasing how different the within-scanner from between-scanner repeats are for each participant. Fig3b shows median correlation values for session pairs for between subject/same scanner, between-scanner/same subject and within-scanner/same-subject. Between-subject variability has little overlap with scan-rescan variability of a subject, however it overlaps quite substantially with between-scanner variability. Fig3c compares IDP-wise between-scanner coefficients of variation (CoVs) to baselines of within-scanner and between-subject variability. We plot IDP-wise and IDP group-averaged relative differences (middle plot) and find that the between-scanner variability can be 2-5 times more than within-scanner. Similarly, between-scanner variability is compared to between-subject variability, showcasing that the former be as large as and occasionally exceed the latter.
We explored how ON-Harmony can be used to assess harmonisation efficacy, e.g. ComBat [7], [16] (explicit harmonisation). Pre/post harmonisation between-scanner variability was compared to within-scanner variability (Fig4a) for example IDP groups. Reductions in between-scanner variability following harmonisation were revealed but did not match the within-scanner baseline. ON-Harmony can also be used to assess the performance of pipelines/tools across scanners (implicit harmonisation). Fig4b shows the results of white matter tractography (FSL-XTRACT [17]). For each scanner, subject-averaged tract maps were obtained and correlated against an atlas, with moderate-high correlation values across all scanners and generally consistent trends across scanners and phases, although with some exceptions. Such comparisons showcase how ON-Harmony can be used to assess susceptibility of tools/pipelines to between-scanner effects. We have presented a comprehensive harmonisation resource (ON-Harmony) for multimodal neuroimaging data, based on a travelling-heads paradigm. ON-Harmony is openly released and may be used to assess harmonisation efficacy, neuroimage processing pipelines/tools and to develop new vendor agnostic tools.
Shaun WARRINGTON (Nottingham, United Kingdom), Andrea TORCHI, Olivier MOUGIN, Jon CAMPBELL, Asante NTATA, Martin CRAIG, Stephania ASSIMOPOULOS, Fidel ALFARO-ALMAGRO, Stephen M SMITH, Adam J LEWANDOWSKI, Karla L MILLER, Mark JENKINSON, Paul S MORGAN, Stamatios SOTIROPOULOS
16:30 - 16:40
#47959 - PG013 Combining single and double diffusion encoding for improved dendritic spine density estimation with magnetic resonance spectroscopy.
PG013 Combining single and double diffusion encoding for improved dendritic spine density estimation with magnetic resonance spectroscopy.
Diffusion-weighted MR spectroscopy (dMRS) probes brain microstructure by examining cell-specific metabolite diffusion, such as N-acetylaspartate (NAA), predominantly found in neurons.
Recent works investigated the sensitivity of Single Diffusion Encoding (SDE)[1-3] and Double Diffusion Encoding (DDE)[4-6] to fine cellular structures, such as dendritic spines.
Here, we consider two acquisition sequences: SDE with high b-values and DDE in the long mixing time regime, where sensitivity to soma size, branching and dendritic length is minimal[4]. Our goal is to evaluate the robustness of spine density estimates in realistic scenarios using those two acquisitions, and how combining them can help reducing the estimates bias, uncertainty and degeneracies.
Using a gray matter (GM) model of spiny dendrites and somas, we simulate SDE/DDE signals, estimate spine density posterior distributions, and compare with in-vivo dMRS mouse data.
Acquisition sequences (matched for simulations and in-vivo data):
SDE: b-values=[0.02,0.5,1.5,3,6,10,15,20]ms/µm^2, 16 diffusion encoding directions uniformly distributed per b-shell, diffusion time Δ=54.2ms, gradient duration δ=3.1ms, TE=58.4ms.
DDE: Δ=30ms, δ=4.5ms, mixing time=29.5ms, b-values=[1, 7.5]ms/µm² per diffusion block, TE=144ms, 32 gradient directions isotropically distributed on a half-sphere for the first block and second block’s orientation w.r.t. the first block θ varying from 0° to 180° in 45° steps. Reported signals are averaged across the 32 directions.
Numerical Simulations (Fig.1):
Model 1: Isotropically distributed spiny dendrites were modeled with the Trees-Toolbox[7] for skeleton building and meshed in Blender. We simulated the SDE/DDE signals using the Monte-Carlo simulator DisimPy[8] (10^6 spins, 9402 time steps, periodic boundary conditions), applying 16 isotropic rotations to minimize macroscopic anisotropy bias. Spine densities σ ranged from 0 to 3.5 spines/µm and diffusivities D from 0.25 to 0.45 µm²/ms, reflecting metabolite diffusivities.
Model 2: Adds a non-exchanging spherical compartment (radius=5µm, same diffusivity as spiny dendrites) to Model 1 to represent somas in GM[9]. Signals were generated analytically using MISST[10], with soma signal fractions fs∈[0.05;0.34].
In-vivo data: Seven C57BL/6J mice (4 overexpressing amyloid precursor protein, APP+; 3 wild-type, APP-) were scanned on a 11.7T scanner using a cryoprobe.
The MRS acquisitions were performed in a 15 μL voxel positioned in the cortex, using the protocols in [11] (SDE) and [5] (DDE). Water signal was suppressed using a VAPOR module. Signal post-processing was performed as described in [12].
Inference: We estimated posterior distributions of σ, D and fs using µGUIDE[13], a Bayesian inference framework, using the SDE and DDE signals separately or combining them. We extracted three quantities from the posterior distributions: the maximum-a-posteriori (MAP), an uncertainty value, based on the interquartile range, and assessed degeneracies, that is multimodality in the distributions. We trained it on 2x10^5 simulations with random combinations of the models parameters uniformly sampled from biologically plausible ranges, with added Gaussian noise with SNR~N(75,7.5) or SNR~N(135,13.5) to match real data, which respectively corresponds to the noise in the acquired tNAA individual mouse signals and in the group-averaged signals. Fig1d-e show examples of SDE and DDE normalized signals for varying spine densities. SDE signals attenuate less with increasing spine density, while DDE θ-modulation decreases, suggesting both sequences are sensitive to spine density.
To characterize the SDE signals, we fitted a bi-exponential model f*e^(-b*D1)+(1-f)*e^(-b*D2) to synthetic signals. Fig.2 shows how D1, D2 and f vary with spine density, without and with soma, with trends preserved under noise. For DDE, a cosine fit A+Bcos(2θ) confirmed strong dependence on spine density, as presented in [6].
Fig.3 presents the estimated MAPs versus ground truth values used for simulating the signals, uncertainty values distribution, and the percentage of degenerate posterior distributions when considering SDE and DDE signals separately or jointly.
Finally, µGUIDE was applied to group-averaged (APP– and APP+) and individual mice in-vivo signals (Fig.4). Fig.4b (Model 1) shows distinct posterior distributions between the two groups, suggesting measurable differences in microstructure. Combining SDE/DDE acquisitions allows to reduce uncertainty and remove almost all degeneracies.
The APP mouse model is known to lead up to 50% decrease in spine density and plaque-associated dystrophic neurites with disrupted trajectories[14], which agrees with our estimates of σ and D from the neuronal tNAA. Using simulations, in-vivo mouse data and AI-based Bayesian inference, we show that combining SDE and DDE-MRS enables accurate and precise estimation of dendritic spine density, providing a new avenue for in-vivo studies of brain GM microstructure.
Maëliss JALLAIS (Cardiff, United Kingdom), Sophie MALAQUIN, Kadir SIMSEK, Julien VALETTE, Marco PALOMBO
16:40 - 16:50
#47764 - PG014 Uncertainty Quantification in SPICE Reconstruction of MRSI.
PG014 Uncertainty Quantification in SPICE Reconstruction of MRSI.
SPICE (Spectroscopic Imaging by exploiting Spatiospectral CorrElation) is an emerging low-rank reconstruction approach for Magnetic Resonance Spectroscopic Imaging (MRSI) [1-4]. It leverages partial separability [5,6] through decomposing the MRSI data into the product of the metabolites’ spatial distribution coefficients U and temporal basis V. Recent advances in subspace learning [2] allows V to be preset by combining empirical measures of metabolite concentrations and other model parameters (e.g. linewidths) with quantum simulations of the spectral response, eliminating the need to jointly estimate both U and V. Moreover, incorporating anatomical priors further reduces noise [9]. SPICE has achieved significantly higher resolution, and faster MRSI, compared with other multi-voxel (MRSI) techniques, e.g., CSI and EPSI [4].
While SPICE has shown transformative potential [4,7,8], its voxel-wise variance in metabolite concentration estimates (i.e. “uncertainty”) derived from spectral fitting of SPICE-reconstructed MRSI data remains underexplored. In particular, the impact of anatomically derived spatial regularisation has been largely overlooked in existing variance analyses [4].
This study aims to quantitatively investigate SPICE uncertainty by comparing: (i) between direct Fourier Transform (DFT) and SPICE; (ii) the effect of spatial constraints on SPICE; (iii) and different undersampling conditions.
Data Generation:
We synthesized a 1D anatomical phantom—a single line of voxels from a 2D MRSI phantom—using the BART brain template [11], which also served as the anatomical prior for generating a spatial constraint (regularisation). Glutamate (Glu) and choline (Cho) concentrations were set to different values in white matter (WM) and gray matter (GM), with all other metabolites assumed as constant background [12]. The ground truth metabolites concentration was defined on the phantom such that Glu exhibits higher concentrations in GM, Cho is elevated in WM, and both are low in cerebrospinal fluid.
Overall Method:
SPICE reconstruction (Fig. 1) consists of optimising U under spatial constraints controlled by lambda (relative weighting of constraints) and Dw (with parameter Wmax controlling the structure of the priors). The optimal U is then multiplied with the pre-trained temporal basis V to obtain spatiotemporal data. However, V is not directly interpretable as physically meaningful metabolite spectra, and no direct transformation exists. Therefore, a spectral fitting process using FSL-MRS [10] was applied to the SPICE-reconstructed spatiotemporal data to estimate metabolite concentrations.
To assess uncertainty, we approximated the covariance of the SPICE-estimated spatial coefficients using the Laplace approximation. This covariance was used to generate Gaussian-distributed samples around the mean U, which were then multiplied by V to synthesize spatiotemporal data. These synthetic datasets were further processed with spectral fitting, providing an efficient Monte Carlo (MC)-style uncertainty estimate.
A further MC simulation was conducted to validate this uncertainty estimation approach. We fixed the SNR and generated multiple k-t space datasets by adding standard normal noise. These datasets were processed through the SPICE + fitting pipeline repeatedly to approximate the uncertainty in metabolite concentrations.
Undersampling was achieved by randomly removing 25 or 50 % of k-space points in each MC repetition. Compared with DFT-based reconstruction, SPICE exhibits a non-uniform uncertainty distribution in spectral domain data. Incorporating spatial priors into SPICE reduces the overall uncertainty in the fitted metabolites (Fig. 2), but uncertainty varies spatially depending on the anatomical reference used as a constraint.
For spatial constraints, as illustrated in Fig. 3., λ increase dampens overall fluctuation, while a larger Dw sharpens contrast between constrained and unconstrained regions. However, stronger spatial constraints increase bias compared to ground truth. Incorporating anatomical priors significantly affects the spatial pattern of uncertainty. Specifically, elevated uncertainty is observed near anatomical boundaries (also in Fig 2.). This can be attributed to the masking matrix Dw [9] allowing edge flexibility while enforcing smoothness in homogeneous regions.
As shown in Fig. 4, undersampling increases the overall uncertainty and bias while roughly preserving the spatial distribution of both measures. This study uses the Laplace approximation and Monte Carlo simulation to quantify the voxel-wise uncertainty and bias in metabolite concentration estimates derived from spectral fitting of SPICE-reconstructed MRSI data. The results demonstrate that incorporating anatomical prior information leads to a non-uniform uncertainty distribution, with elevated uncertainty observed near tissue boundaries. Propagation of uncertainty through SPICE-like reconstruction and fitting is shown to be possible.
Tian LYU (Oxford, United Kingdom), Simon M FINNEY, Saad JBABDI, William T CLARKE
16:50 - 17:00
#46299 - PG015 Quantification of the Dipolar Order Relaxation Time (T1D) Combining Solid-State NMR and ihMT MRI.
PG015 Quantification of the Dipolar Order Relaxation Time (T1D) Combining Solid-State NMR and ihMT MRI.
Inhomogeneous magnetization transfer (ihMT) imaging has been proposed as a novel myelin sensitive contrast mechanism [1]. IhMT requires the presence of residual dipolar couplings and is mainly driven by the dipolar order relaxation time (T1D) [2,3] motivating a detailed quantitative assessment. However, the heterogeneity of biological tissues implies a challenge. There is still no consensus in the scientific community on how to measure T1D for systems composed of 'mobile' and 'solid' protons. Instead, techniques from solid-state NMR such as adiabatic demagnetization and remagnetization in the rotating image (ADRF/ARRF) or the Jeener-Broaekaert (JB) [4] sequence were used to quantify T1D [2,5-9] - both developed for the study of ‘pure solids’ Alternatively, an assessment of T1D has been proposed using a modified ihMT sequence by varying the filter time (τswitch) between RF pulses applied at positive and negative offset frequencies (ΔRF), resulting in a modification of the ‘dual-sided’ saturation scheme [9,10].
Using distinct methods hinders comparisons of T1D estimates, because of variations in sequence implementation, data processing, and analysis. To date, only a few studies have compared the relaxation of dipolar order using distinct techniques [5, 8].
Hair conditioner (‘ihMT phantom’ [10,11]) was investigated with a 500 MHz NMR spectrometer (BRUKER AVL500 equipped with a BBFO+ probe, 90° pulse duration ≈13.7 μs, TE=6.5 μs, TopSpin 3.6.2) at ≈27° without using the deuterium lock. For comparison, the characterization of T1D was carried out using NMR and ihMT MRI techniques.
For the JB experiments, two implementations (Fig. 1A,B) were compared (equal number of 32 scans): The ‘standard’ JB sequence [6], and an extended version with an additional refocussing RF pulse to compensate for off-resonant signal contributions due to field inhomogeneities or chemical shifts. The ADRF measurements (Fig. 1C) were performed with a ramp time (τramp) of either 1 or 4 ms [2,5].
The ‘τswitch’ experiments mimic the approach utilized in MRI studies [9,10]. Briefly, a train of RF pulses (Gaussian, truncation level 1%) with a constant number of RF events but a different number of consecutive pulses of identical offset (Fig. 1D) was used to create sensitivity to T1D. Experimental analyses were conducted using a simplified two-pool model (2PM) equipped with a dipolar reservoir and an established matrix-algebra approach [12-14] to estimate the biophysical model parameters, including T1D. JB analysis for systems containing aqueous and non-aqueous protons, like hair conditioner, is surprisingly complex (Fig. 2). At a first sight, the analysis of the magnitude spectra reveals (at least) two distinct dipolar relaxation times. However, the long apparent T1D is associated with the relaxation of remaining (mainly off-resonant) ‘Zeeman’ signal contributions, as confirmed by simulations (not shown here). These confounding signal contributions are removed more efficiently with the extended JB sequences due to the additional 180° pulse, making the fit of dipolar order decay more reliable. A sufficient description of the decay of the ‘positive lobe’ signal can be achieved with a mono-exponential fit, resulting in a T1D estimate of ≈11 ms. The first τ-values were always excluded from the analysis to avoid contamination of the dipolar decay with the proton solid echo [2] and to give the system time to reach a new equilibrium. The analysis of the ADRF/ARRF experiments was performed in an analogous manner but must be considered less reliable as water signal contributions strongly dominate the detected signal. However, the estimated T1D times (e.g., ≈15 ms for the measurement with τramp=1 ms) were in the same order of magnitude.
The results of the 2PM analysis of the ihMTR as a function of τswitch are shown in Fig. 4. Since the observed signal decay is mostly driven by T1D several model parameters were fixed (T1A = 2s, T1B = 0.5s). Estimated T1D values (≈9-16 ms) with the 2PM agree surprisingly well with JB analysis, where exchange processes of non-aqueous protons with aqueous protons are not considered and the dipolar order decay is directly accessed. Noticeable deviations in the fit were observed for highest B1+-amplitudes (strongly exceeding clinical B1,RMS≈3 μT [1]) , which may result from the manifestation of another ‘short’ T1D component as previously reported [1, 5]. However, it must be stressed out that the requirement of the Provotorov theory (ω1 << γHloc [15], e.g. 23.2 kHz*2π for H-H with r=1.73Å) is potentially violated (for 85.6 μT, ω1 ≈ 3.6 kHz*2π). Reliable quantification of T1D in biological systems is a challenging but essential task to exploit the full potential of T1D contrast as a biomarker of myelin. For the first time, a good agreement of T1D estimates has been achieved using solid-state NMR and ihMT MRI - an important step towards a better understanding of dipolar order-related contrast in MRI.
Niklas WALLSTEIN (Marseille), Pierre THUREAU, André PAMPEL, Olivier M. GIRARD, Axelle GRÉLARD, Lucas SOUSTELLE, Erick DUFOURC, Guillaume DUHAMEL
|
Auditorium 900 |
|
"Thursday 09 October"
Added to your list of favorites
Deleted from your list of favorites
B12
15:30 - 17:00
ET2-1 - AI in MRI
ET2: Cycle of Clinical Practice
15:30 - 17:00
AI in Body.
Laure FOURNIER (Keynote Speaker, Paris, France)
15:30 - 17:00
AI in Cardiac.
Alexis JACQUIER (Chef de service) (Keynote Speaker, Marseille, France)
15:30 - 17:00
AI in MSK.
Thomas VAN DEN BERGHE (Keynote Speaker, France)
15:30 - 17:00
AI in Neuro.
Christian FEDERAU (Keynote Speaker, Switzerland)
15:30 - 17:00
Explainable MRI.
Jonas TEUWEN (Keynote Speaker, The Netherlands)
15:30 - 17:00
MSK MRI before and after AI - a radiographer's perspective.
Alibhe DOHERTY (Keynote Speaker, Ireland)
|
Espace Vieux-Port |
|
"Thursday 09 October"
Added to your list of favorites
Deleted from your list of favorites
C12
15:30 - 17:00
FT2 LT - Translational MRI: from metabolism to therapy
FT2: Cycle of Translation
15:30 - 15:32
#47879 - PG097 Assessment of Deep-Learning Reconstruction Algorithms for Preclinical MRI.
PG097 Assessment of Deep-Learning Reconstruction Algorithms for Preclinical MRI.
Deep-learning reconstruction algorithms have been recently proposed to reduce the prolonged scan times in MRI exams [1-3]. Recent deep-learning methods employ neural networks for MRI reconstruction by learning data-driven image priors. Various neural network architectures, including U-Net [4,5], generative adversarial networks (GAN) [6], and transformers [7], have been used for deep MRI reconstruction. However, they have been applied mostly to clinical MRI in the literature, and deep MRI reconstruction has not been fully explored for preclinical MRI scans. Preclinical MRI exams concern imaging of substantially smaller structures at higher spatial resolutions and smaller field-of-view, along with differences in the imaged anatomy between species. The main purpose of this work is to apply several deep learning-based accelerated MRI techniques, including U-Net [8], CascadeNet [9], PDNet [10], DiffuseRecon [11], and TC-DiffRecon [12], to reduce the scan time of the preclinical anatomical MRI and to evaluate their respective performances.
This study utilized two different rat brain MRI datasets: an open-access dataset [13] and an in-house dataset. The open-access dataset was collected at the Center for Animal MRI (CAMRI) at the University of North Carolina. This dataset includes 132 adult male rats, with T2-weighted rapid acquisition with relaxation enhancement (RARE) and T2*-weighted echo planar imaging scans acquired on a 9.4T MRI scanner [14]. Only the RARE images were used in this study, comprising 5,172 slices. The in-house dataset was acquired on a 7T preclinical MRI scanner (MR Solutions, UK) and included T1-weighted Fast Spin Echo (FSE) (TR/TE = 1193/11 ms, flip angle = 90°, matrix = 256x238, FOV = 2.5 cm) and T2-weighted FSE (TR/TE = 5000/45 ms, flip angle = 90°, matrix = 256x238, FOV = 2.5 cm) sequences. Animal MRI data were retrospectively undersampled across the phase-encoding dimension to achieve an acceleration factor of R=4, using a 1D uniform-density random mask with an 8% calibration region (Figure 1).
Five deep learning-based MRI acceleration methods were compared in this study, including data-driven and physics-driven methods (U-Net, CascadeNet, PDNet) and also two generative diffusion methods (DiffuseRecon and TC-DiffRecon). Furthermore, L1 wavelet regularization-based reconstruction was applied as the CS method by using the BART [15] framework for comparison purposes. The performances of these MRI reconstruction algorithms were evaluated using peak signal-to-noise ratio (PSNR) and structural similarity index (SSIM). Figures 2 and 3 show the results of the accelerated MRI reconstruction techniques on CAMRI and the in-house MRI datasets, respectively. The first column represents the ground truth rat brain MRI and zero-filled reconstructed image. Reconstructed images and corresponding average metrics for each deep learning-based accelerated MRI technique are also shown. The CS method improved the image reconstruction compared to the zero-filled Fourier reconstruction but performed worse than the deep learning-based methods on both datasets. Among the deep learning algorithms, PDNet achieved the best reconstruction performance with an SSIM and PSNR of 0.912 and 36.731dB, respectively, on the in-house MRI dataset. However, generative diffusion models (DiffuseRecon and TC-DiffRecon) underperformed compared to data-driven and physics-driven techniques with an SSIM of 0.798 and 0.822, respectively. This initial study investigated the performance of several deep learning-based acceleration techniques on rat brain anatomical MRI. Our results indicated that generative diffusion models performed worse than data-driven and physics-driven accelerated MRI techniques, in contrast to the promising outcomes observed on human MRI datasets—a finding that warrants further investigation. Additionally, using pre-trained models from large human MRI datasets could enhance reconstruction performance and improve generalization capability in preclinical studies. The preclinical MRI acquisitions could be accelerated by using deep learning-based reconstruction techniques, which would be beneficial in reducing scan times and animal anesthesia duration. Physics-driven accelerated MRI techniques achieved best reconstruction performance compared to data-driven and generative diffusion models in terms of SSIM and PSNR on two different rat brain anatomical MRI datasets.
Said ALDEMIR (Istanbul, Turkey), Oguzhan HURAYDIN, Uluc PAMUK, Pinar S. OZBAY, Tolga CUKUR, Esin OZTURK-ISIK
15:32 - 15:34
#47584 - PG098 A method for MRI-Only interstitial needle reconstruction of the Syed-Neblett applicator in gynecological brachytherapy.
PG098 A method for MRI-Only interstitial needle reconstruction of the Syed-Neblett applicator in gynecological brachytherapy.
Gynecological brachytherapy (BT) treatment planning has conventionally relied on CT scans to visualize and reconstruct applicators and needles, while MRI has been primarily used for delineating anatomical structures. However, some institutions have moved toward MRI-only planning approaches. An MRI-only approach eliminates registration errors between CT and MRI. It also reduces the time of the entire procedure, minimizing errors from motion and anatomical changes due to a longer procedure process, and improving the patient experience. Planning based solely on MRI presents certain difficulties, as the modality is prone to image distortions, which can impact dose distribution. Moreover, needle reconstruction can be more difficult due to the lack of options for MRI needle markers. A commercial MRI line marker exists; however, it is not approved for clinical use in many countries outside the United States. The Syed-Neblett template interstitial applicator (Best Medical) is a typical applicator choice for treating gynecological cancers that extend beyond 5 cm in lateral width. It is depicted in Figure 1. It consists of a central tandem which enables inserting a single needle through its inner cylinder, and 6 peripheral needles along the white plastic edge, with the addition of a groove to mark the orientation. Additional outer needles can be inserted using the purple template indicated. The use of this applicator presents challenges in MRI reconstructions: the 6 peripheral needles on the central tandem are not distinguishable from the surrounding plastic on MRI. Moreover, the central needle may be surrounded by void in a patient, and is thus also potentially not visualized in MRI images. We herein present a solution for needle reconstruction on MRI using this applicator without the need for MRI needle markers.
A library model of the central cylinder was designed in MIM (Medical Image Merge, version 7.2.8) using a CT image of the cylindrical white tandem. CT-visible needle markers were used to define the position of the 6 peripheral needles relative to the plastic cylinder and its groove. The contour in MIM was fixed in shape and size, allowing only translation and rotation to align with MRI images. This model thus serves as a guide for reconstructing the 6 peripheral needle paths in the cylinder on MRI images, without needle markers. Needle reconstruction was done in Oncentra Treatment Planning system (Elekta, version, 4.6.3), upon import of the cylinder model and MRI images. The inferior needle location in the model, along with the visible superior location where needles exit the cylinder into gel or tissue, enables full reconstruction. An end-to-end test using a gel phantom mimicking the MRI properties of water was conducted with the Syed-Neblett applicator. Fifteen needles were inserted—six in the central cylinder and nine in the outer template. CT and MRI images were acquired. Figure 2 shows an axial MRI with the cylinder model (blue outline) overlaid to indicate the indistinct peripheral needles. Alignment was done using the central groove. Reconstruction was completed on both CT and MRI images, and the CT-based plan was transferred to the MRI reconstruction. Dose grids were calculated for CT and MRI reconstructions, and a gamma analysis (3%/2mm, 10% dose threshold) was completed in MIM to compare the CT and MRI dose maps. For the unidentifiable central needle, a clinical solution is to fill the tandem center with gel and gauze, permitting visualization on MRI. If not visible, misplacement was simulated by modeling the needle at opposite ends of the 7 mm inner cylinder to represent a worst-case scenario. The needle’s exit point remains clearly visualized. A gamma analysis was completed to compare these 2 modelled cases. In this test, the central cylinder needles were conservatively loaded with 40% of the total dwell time to simulate a plan heavily weighted to the central region. The gamma analysis for the dose grids calculated on the CT and the MRI plan yielded a result of 97.6 %. The gamma analysis for the dose grids obtained modelling the central needle at worst-case opposite ends of the central tandem was 99.92 %. The gamma analysis results indicate that using the developed model in an MRI-only gynecological brachytherapy reconstruction results in a clinically equivalent dose distribution compared to using a CT reconstruction, within established tolerances. The central needle in the central cylinder can be visualized by applying medical gel in the inner cylinder itself. The dosimetric error associated with mis-constructing this central needle is minimal, with a gamma analysis indicating a pass rate of 99.92% for a modelled worst-case scenario. The method presented herein describes a possible workflow for needle reconstruction using the Syed Neblett template in an MRI-only gynecological brachytherapy treatment, without the use of MRI needle markers.
Clara J FALLONE (Calgary, Canada), Shima Y TARI, Philip MCGEACHY, Tyler MEYER
15:34 - 15:36
#47627 - PG099 Extensive experimental validation through MRI thermometry of hybrid model for bioheat transfer in microwave ablation of liver and reconstruction of the temperature field near the needle.
PG099 Extensive experimental validation through MRI thermometry of hybrid model for bioheat transfer in microwave ablation of liver and reconstruction of the temperature field near the needle.
Gas-induced MRI artifacts may occur during microwave ablation (MWA) due to extreme heating temperature. First described by Viallon et al. [Ref1], these gas formations significantly distort the measurements, in approximately half of the cases, by introducing a characteristic imaging artifact (dipole). This issue could be overcome through continuous integration of the temperature field from simulation of the surrounding tissue. This work aims to validate on ex-vivo cow liver tissue data (N=16) the use of a hybrid model designed to calculate the temperature field during MWA procedures. The second purpose of this work is to reconstruct the temperature field near the needle where boiling artifacts occur based on the calibrated simulation on in-vivo pig liver data(N=2).
An AveCure microwave system (MedWave, San Diego, USA) was used to perform the ablation (in ex-vivo bovine liver tissue) using a 14-gauge large antenna inserted percutaneous under MRI guidance. The MRI-compatible device was connected to a generator located inside the Faraday cage using a shielded cable provided by the manufacturer. Ablation duration was set to 5/10/7’30/15min with a target temperature of 60/80/100/120°C at room temperature. MR temperature images were obtained on a 1.5 T clinical MRI (Magnetom Sola Fit, Siemens Healthineers) using a multi-slice multi-shot echo planar imaging (EPI) sequence positioned parallel to the microwave probe. A stack of 13 slices was acquired dynamically every 4 s using the following parameters: TE=19 ms, TR=47.29 ms, FA = 40°, FOV was 300×300 mm², matrix size= 128×128, in plane spacing = 2.34×2.34 mm², slice thickness = 3 mm, slice gap = 1.5 mm, bandwidth/pixel = 953 Hz, GRAPPA acceleration factor=2.
In-vivo MWA were performed using the same device in swine liver during 7’30 min. Acquisitions were performed under respiratory gating using the most stable part of the expiration using the same parameters with 7 slices. Temperature calculation was carried out employing the Proton Resonance Frequency Shift method.
The proposed model is based on the resolution of the bioheat equation under spherical symmetry (see fig.1). By applying changes of variables and the numerical inversion of the Laplace transform using Stehfest’s algorithm [Ref 2] combined with finite difference method, the voxel-averaged temperature is computed by integrating the temperature distribution over a 1mm3 isotropic grid representing the voxel network. The central circumference of the spherical model is repeated and incorporated into a hemisphere-based model to approximate the elongated shape of the temperature field. The regridding to experimental voxel size and realignment of the simulated data with the experimental MRI data is done by using ANTs [Ref 3]. The optimization is performed using least squares algorithm[Ref 4]. The refined simulation refers to the simulation using the optimized parameters on a 1mm3 isotropic grid, realigned to match the experimental data. The model estimates heat source parameters Q0 and β (equation figure 1) while assuming constant thermal properties. The average of the estimated parameters on 5/7’30/10 min have been taken to compute the 15 min cases (Cross Validation). Table 1 lists the parameters Q0 and β that were estimated from ex-vivo data with corresponding RMSE. The 100/120°C cases have a higher RMSE because of the influence of the elevated noise. Cases 80/60°C indicate RMSE of 6.5°C and 12.1°C respectively. Temperature maps of the experimental (ex-vivo) and simulated data (before and after optimization) are displayed in figure 2. One can see the simulation accurately represents the temperature distribution over time. Temperature maps of the experimental (in-vivo) and simulated data (before and after refinement) are displayed in figure 3. It allows to predict the variation (in time and space) of the ablation while artifacts caused by boiling. In both cases, the artifact appears at repetition 70 and 90 respectively, which corresponds to a temperature of 86°C and 91 °C in the refined simulation. The grid refinement allows to some extent overcome the boiling artifact and might provide data closer to the real maximum temperature. Nonetheless, the values found are lower than 100°C, thus tests on data sets at finer grids (such as 500 µm3 isotropic grid) are upcoming to try to validate this statement. Yet the computational cost remains excessive for a real-time estimation, this is one of the current goals. Also, the optimization process needs to be improved in order to obtain an even lower RMSE. This model is a promising candidate for real-time estimation during MWA procedures for liver tumour. It offers accurate temperature reconstruction and allows to estimate the parameters of the heat source while keeping an acceptable RMSE. The work conducted with in-vivo data showed one could estimate temperature when a boiling artifact occurs. Future work will focus on extending the current approach to clinical patient cases.
Ida BURGERS (Bordeaux), Mariana DE MELO ANTUNES, Nino AVETIKOVI, Luigi NARDONE, Bruno QUESSON, Pierre BOUR, Thibaut FALLER, Max SEIDENSTICKER, Olaf DIETRICH, Jean-Luc BATTAGLIA, Valery OZENNE
15:36 - 15:38
#47079 - PG100 Magnetic resonance thermometry validation of a bioheat transfer model for lesion size prediction in liver tumor microwave ablation procedures.
PG100 Magnetic resonance thermometry validation of a bioheat transfer model for lesion size prediction in liver tumor microwave ablation procedures.
Precise heat delivery is essential in microwave liver tumor ablation to ensure complete tumor destruction while sparing healthy tissue. Computational models can aid treatment planning and outcome prediction, but their clinical relevance is limited without validation on experimental data. Magnetic resonance thermometry (MRT) offers a valuable reference for such validation. This study aims to validate a COMSOL Multiphysics model of microwave ablation using MRT data acquired in ex vivo cow liver (N=12). The model was further evaluated in vivo by comparing simulated and observed lesion sizes in pig liver (N=1).
Experimental data description: An AveCure microwave system (MedWave, San Diego, USA) was used to perform the ablation using a 14-gauge large antenna inserted percutaneously under MRI guidance. Ex vivo experiments were conducted under varying conditions: ablation duration of (5, 7.5, 10, 15 min) and target temperatures of (60°C, 80°C, 100°C). MR thermometry was performed based on the PRF method and images were obtained on a 1.5 T clinical MRI (Magnetom SolaFit, Siemens Healthineers) using a multi-slice multi-shot echo planar imaging (EPI) sequence positioned parallel to the MW probe. A stack of 13 slices was acquired dynamically every 4 s using the following parameters: TE=19 ms, TR=47.29 ms, FA = 90°, FOV was 300 mm times 300 mm, matrix size= 128x128, in plane spacing = 2.3mm times 2.3 mm, slice thickness = 3 mm, slice gap = 1.5 mm, bandwidth/pixel = 953 Hz, GRAPPA acceleration factor = 2. In vivo acquisitions were performed under respiratory gating during the most stable part of exhalation. At the end of the experiment, gadoteric acid (0.5 mmol/kg, GE Healthcare) was injected intravenously, and 3D T1-weighted images were acquired to visualize the non-perfused volumes at a resolution of 1.2×1.2×1.2 mm³. Simulations: Finite element simulations were conducted using COMSOL Multiphysics (version 6.3, COMSOL Inc.). The simulation modeled the coupling between the electromagnetic field and bioheat transfer, representing the interaction between microwave energy deposition and tissue heating via the Pennes bioheat equation. To reduce computational complexity and runtime, a 2D axisymmetric model was employed, assuming a homogeneous and symmetric tissue environment. The 2D temperature map was revolved around the symmetry axis to generate a 3D temperature distribution. This 3D data was then voxelized into a 4D matrix, where each element represents the average temperature within a 1 mm³ voxel at a specific spatial location and time point (Fig.1). Optimization and cross validation: A constrained optimization pipeline was developed based on a grid search approach to minimize the mean squared error (MSE) between simulated and experimentally measured temperature distributions. A mask was generated by combining two criteria: the first excluded voxels with excessive noise, and the second retained only the voxels located within the heating region. The MSE was then computed over the retained voxels. The optimization focused on two key parameters: the slot position along the microwave ablation antenna (from 20 to 42 mm) and the input power level (between 10 and 32 W). A 7×7 optimization grid was used for parameter space exploration. For each setpoint temperature, optimal parameters were identified for 5, 7.5, and 10-minute heating durations. The averaged parameters were then used to simulate the 15-minute heating, serving as a cross-validation step. (Fig.1) Lesion size estimation: In addition, the COMSOL model was validated using in vivo data by comparing simulated and experimental lesion sizes. Thermal dose was estimated using the CEM43 model, and experimental lesions were segmented from post-ablation T1-weighted images enhanced with gadolinium. The Dice similarity coefficient was used to quantify the spatial overlap between simulated and observed lesions. For the 60°C and 80°C datasets, the optimization process yielded a root mean square error (RMSE) ranging from 6 to 12°C (Fig.2-3). In contrast, the 100°C dataset showed higher errors of 23°C on average, likely due to reduced reliability of the experimental data, as boiling-induced susceptibility artifacts significantly distort the MR signal at high heating regimes. The in-vivo comparison between experimental and simulated lesions resulted in a Dice similarity coefficient of 0.73 (Fig.4). This study involved several simplifications: tissue properties were assumed constant, a 2D axisymmetric geometry was used, and the exact antenna design and feedback mechanism were not modeled due to limited specifications. Liver anatomy and vessel structures were also excluded. While these factors limit accuracy, the model still showed good agreement with experimental data, indicating strong predictive potential. Future improvements should focus on incorporating temperature-dependent properties and anatomical detail. In the near future the presented work will be extended to clinical data. None
Nino AVETIKOVI (Bordeaux), Ida BURGERS, Luigi NARDONE, Pierre BOUR, Thibaut FALLER, Bruno QUESSON, Olaf DIETRICH, Max SEIDENSTICKER, Jean-Luc BATTAGLIA, Valéry OZENNE
15:38 - 15:40
#47873 - PG101 Repeatability analysis of DCE-MRI reveals heterogeneous responses to anti-vascular therapy in patients with metastatic colorectal cancer.
PG101 Repeatability analysis of DCE-MRI reveals heterogeneous responses to anti-vascular therapy in patients with metastatic colorectal cancer.
Functional imaging is used widely to detect, quantify and track changes in the tumour microenvironment induced chemotherapy, radiotherapy, targeted therapies and immunotherapy. One leading technique is dynamic contrast-enhanced MRI (DCE-MRI), where biomarkers such as median Ktrans have quantified anti-vascular drug effects in around 80 early phase clinical trials.
Investigators typically report cohort-level effects (e.g. reduction in median Ktrans to indicate reduction in blood flow and/or permeability). Although useful, this approach does not necessarily consider the variety of within-patient and between-patient physiologic changes occurring.
Here, we investigate the relative merits of two approaches that aim to capture the heterogeneity of vascular responses induced by an anti-angiogenic drug. Initially we examine latent class mixed modelling (LCMM). This assumes that the patient population is heterogeneous and consists of multiple latent classes, each characterized by distinct median Ktrans trajectories. As an alternative, we impose ‘vascular response’ classes on each tumour depending on the magnitude of change in median Ktrans, relative to test-retest precision of the biomarker.
Retrospective analysis was performed on data from patients with metastatic colorectal cancer recruited for a clinical trial. All patients had two DCE-MRI scans pre-treatment, before one cycle of 10mg/kg bevacizumab monotherapy during which further scans were acquired at day 2 (d2) and day 14 (d14; end of cycle 1). Next, patients were scanned at day 21 (d21) halfway through a cycle 2.5mg/kg bevacizumab combined with oxaliplatin/fluorouracil chemotherapy (Figure 1).
Median Ktrans and tumour volume were derived from DCE-MRI performed on a 1.5T Philips machine, using the extended Tofts model and measured input function. Overall cohort effects were measured using mixed effects models on log-transformed data. LCMM used Akaike information criterion (AIC) to define optimal number of classes with distinct trajectories of change in median Ktrans or volume.
Next, individual changes in median Ktrans were identified for each lesion. When the magnitude of Ktrans change exceeded the repeatability coefficient limits of agreement (RC LOA; derived from test-retest pre-treatment scans) this was considered ‘real change’ with 95% confidence and the tumours were considered to be ‘vascular responders’. Similar approaches were used for tumour volume.
All statistics were calculated in RStudio (version 2024.04.0). Significance levels of 0.05 were used, once multiple comparisons were considered. Data from 70 patients were included. Individuals had between 1-6 lesions each (total of 137 lesions). Median Ktrans and tumour volume reduced in the cohort by d2 and persisted to d21 (all p < 0.001). LCMM did not reveal any distinct trajectory profiles, as AIC suggested that one class best fitted the data, implying that changes in median Ktrans and volume were relatively homogeneous across the cohort.
Individual lesion analysis using RC LOA revealed four key findings: (1) individual lesions divided equally into ‘vascular responders’ – where median Ktrans reduction exceeded the RC LOA (n=63-71 d2-21) on at least one of d2, d14 or d21 – compared with ‘vascular non-responders’ (n=62-63 d2-21; Figure 2); (2) when a ‘vascular response’ was observed there was considerable variation in its onset, duration and persistence (also Figure 2); (3) Lesions with ‘vascular response’ at d14 and d21 were more likely to have associated volume decrease than ‘vascular non-responders’ (Chi squared p = 0.0197 at d14 and p = 0.0249 at d21; Table 1); (4) in the 34 patients with multiple lesions, 23 had a discordant mix of ‘vascular response’ and also ‘vascular non-response’ (Figure 3). Cohort analysis identifies the overall trend in vascular or metabolic response to a therapy. This is usually the rationale for including functional imaging into a clinical trial. However, as seen in this study of the anti-vascular agent bevacizumab, techniques designed to identify latent classes can fail to distinguish different response groups.
In distinction, comparing individual lesion changes to test-retest data can identify the variation in response, both in presence or absence and in onset, duration and persistence of effect. Furthermore, patients with multiple lesions more often than not exhibit changes that are complex and discordant. Analysing responses within individual lesions is made possible by using test-retest data. This methodology can reveal extensive within-patient and between-patient heterogeneity of response that is not apparent from the cohort analysis. These findings can have important implications for optimising dose and schedule of drugs in development. More generally, our findings emphasise that current conclusions from functional imaging studies may be overly simplistic.
Nivetha SRIDHARAN, Nuria PORTA, James O'CONNOR (London, United Kingdom)
15:40 - 15:42
#46579 - PG102 Differentiation of tumor progression from pseudoprogression in glioblastoma patients with GRASP DCE-MRI and DSC-MRI.
PG102 Differentiation of tumor progression from pseudoprogression in glioblastoma patients with GRASP DCE-MRI and DSC-MRI.
After surgery and radiochemotherapy (RCT), glioblastoma patients may develop new or enlarging contrast-enhancing lesions indicating either progressive disease (PD) or pseudoprogression (PsP). The aim of this study is to determine the diagnostic accuracy of combined Golden Angle Radial Sparse Parallel Dynamic contrast-enhanced (GRASP DCE-) and Dynamic susceptibility contrast (DSC) MRI in differentiating PD from PsP in glioblastoma patients following RCT.
Retrospective study of glioblastoma patients who had undergone surgery and RCT between 2017 and 2021, developed new or enlarging contrast-enhancing lesions suspicious for PD or PsP, and had GRASP DCE- and DSC-MRI. The diagnostic accuracy of perfusion parameters was evaluated using the Area Under the Receiver Operating Characteristics (ROC) Curve (AUC) to differentiate between PD and PsP at the first suspicion of progression, and at the confirmation scan. Among 83 patients, 62 were classified as PD and 21 as PsP based on lesion outcome observed on serial MRI for all patients, with additional histological confirmation in 18 patients. The median values of the perfusion parameters were generally higher in the PD group in comparison to the PsP group with rCBV (PD: 3.48 [2.49-4.84], PsP: 1.6 [1.33-2.27], p <.001) and Vp (PD: 0.08 [0.06-0.12], PsP: 0.05 [0.04-0.08], p=.032) showing statistically significant differences between groups.
The combination of DSC- and DCE-MRI parameters (rCBV, Ktrans, Vp and Ve) showed a very high AUC of 0.90 (95% CI [0.82, 0.98]) when differentiating PD from PsP at the confirmation scan with the following thresholds: rCBV 2.87 (Sensitivity 71%, Specificity 94%), Ktrans 0.12 (Sensitivity 73%, Specificity 76%), Ve 0.31 (Sensitivity 75%, Specificity 65%) and Vp 0.06 (Sensitivity 79%, Specificity 59%). After completion of postoperative radiochemotherapy, distinguishing progressive disease from pseudoprogression remains challenging on standard sequences, and the current RANO 2.0 criteria struggle to perform this differentiation. This distinction is crucial since PD mistaken for PsP may lead to continuation of an ineffective therapy with consequent tumor growth, while PsP mistaken for PD may lead to discontinuation of effective therapy.
The aim of this study is to determine the diagnostic accuracy of a double perfusion study encompassing Golden Angle Radial Sparse Parallel (GRASP) DCE-MRI combined with DSC-MRI in differentiating PD from PsP in glioblastoma patients following RCT.
The results of the current study show that combining the DSC- and DCE-MRI derived perfusion parameters (rCBV, Ktrans, Vp and Ve) provides an excellent differentiation between PD and PsP with an AUC of 0.90 (95% CI [0.82, 0.98]). Though there was a considerable variation of the sensitivity and specificity of the individual parameters, the following threshold values may assist in distinguishing PD from PsP in clinical practice: rCBV 2.87 (Sensitivity 71%, Specificity 94%), Ktrans 0.12 (Sensitivity 73%, Specificity 76%), Ve 0.31 (Sensitivity 75%, Specificity 65%), and Vp 0.06 (Sensitivity 79%, Specificity 59%).
The strengths of this study are the introduction of the GRASP technique in the assessment of treatment response in glioblastoma alongside DSC-MRI in one single protocol using a split-bolus technique, and the multiparametric analysis of the diagnostic accuracy of combined DSC- and DCE-MRI parameters and their thresholds.
GRASP DCE-MRI is an accelerated acquisition technique that provides high temporal and spatial resolution, enabling accurate estimation of pharmacokinetic parameters in tissues with rapid kinetics, such as glioblastoma . The permeability parameters were measured with a temporal resolution of 4.3 seconds and a spatial resolution of 1.1 x 1.1 x 1.1 mm³, consistent with the QIBA (Quantitative Imaging Biomarkers Alliance profile) recommendations, which suggest a temporal resolution of less than 10 seconds (ideally ≤ 5 seconds) for reliable estimation of Ktrans, Vp, and Ve. Combining GRASP DCE-MRI with the standard DSC-MRI protocol may enhance the differentiation between progressive disease and pseudoprogression in glioblastoma patients following radiochemotherapy. DSC-MRI primarily reflects cerebral blood volume, while DCE-MRI provides complementary information on vascular permeability and plasma volume, allowing for a more comprehensive assessment of tumor physiology. Prior studies have shown that DSC-MRI alone may be limited in cases with blood–brain barrier disruption or susceptibility artifacts. Incorporating DCE-MRI addresses these limitations by introducing additional permeability-based biomarkers. This study also identifies optimal perfusion parameter thresholds from both modalities, supporting more accurate differentiation between PD and PsP. While exploratory, these findings offer meaningful guidance for neuroradiologists in clinical practice.
Alexandra TODEA (Zurich, Switzerland)
15:42 - 15:44
#47896 - PG103 Longitudinal MRI radiomics and MRSI metabolic signatures for progression-free survival prediction in glioblastoma.
PG103 Longitudinal MRI radiomics and MRSI metabolic signatures for progression-free survival prediction in glioblastoma.
Accurate prediction of progression-free survival (PFS) in glioblastoma (GBM) patients during post-surgical follow-up remains challenging. Magnetic resonance spectroscopic imaging (MRSI) captures tumor metabolism, while MRI radiomics quantifies tissue heterogeneity. This paper studies the utility of MRSI features and MRI radiomics combined with clinical data for PFS prediction.
From our prospective clinical trial (NCTxxxxxxxx) [1], we retrospectively analyzed MRI and MRSI data from 74 patients imaged longitudinally after surgical resection and adjuvant therapy until relapse. Clinical data (age, sex, MGMT methylation status, surgery type [biopsy vs. complete resection], radiotherapy treatment [standard vs. boost dose], and corticosteroïd use) were included. Automatic segmentation of the contrast-enhancing (CE) and FLuid-Attenuated Inversion Recovery (FLAIR) hyperintense regions of interest -referred to as CE and Edema-was performed using a nnU-Net neural network [2] trained on post-operative MRI. For each regions of interest, MRSI quantification using a Bayesian model with sparse constraints enabled extraction of 21 metabolites, including both conventional and non-conventional ones [3]. Radiomics analysis [4] derived approximately 1000 features from T1-weighted (pre- and post-contrast), T2-weighted, and FLAIR MRI. Features with >10% outliers were removed, and any pair of features with Pearson |r| > 0.90 was filtered to reduce multicollinearity. The resulting dataset was randomly split into training (70%) and held-out test (30%) sets. Finally, survival analyses employed time-varying Cox proportional hazards models with an L₂ penalty-λ was optimized via 5-fold cross-validation on the training set. For the MRSI model, a univariate time-varying Cox analysis (p < 0.1) identified 13 metabolites, that were combined into an MRSI metabolic risk score. We then refit a multivariable time-varying Cox model including that metabolic score plus clinical covariates. Even after adjustment, the metabolic score remained a significant predictor of progression-free survival (with hazard ratios : Lactate (CE): HR 1.57, 95% CI 1.09–2.26; Glutamine (Edema): HR 1.57, 95% CI 1.15–2.15; see Table 1 and the forest plot in Figure 1). On our held-out test set (Table 2), the MRSI model achieved a C-index of 0.62, time-dependent AUC at 8 months of 0.61, and Uno’s C-index of 0.76.
For the MRI radiomics model, univariate Cox (p < 0.1) retained 168 features. We built a single radiomics risk score from all coefficients (top 10 contributors shown in Table 1) and refit that score plus the same clinical covariates in a multivariable time-varying Cox. The radiomic score was independently prognostic-e.g. GLRLM GreyLevelNonUniformity (FLA-CE) HR 2.82 (95% CI 1.79-4.46), IntensityKurtosis (T1c-Edema) HR 0.61 (95% CI 0.39-0.95) (Table 1, Figure 1) - and on test it achieved C-index 0.74, AUC 0.77, and Uno C-index 0.72 (Table 2).
Finally, when we merged the two risk scores into a single model (Table 2), the C-index (0.69) fell between MRI-only (0.74) and MRSI-only (0.62) performances, reflecting both their overlapping prognostic information and each modality’s unique, complementary signals. These results suggest that MRI radiomic features provide the stronger prognostic signals but they also show that several MRSI‐derived metabolites, including lactate in the contrast enhancing region and glutamine in the edema region, carry relevant information on relapse risk. The consistent significance of glutamine in the peritumoral edema region underlines its potential as a relevant marker of glioblastoma aggressiveness and progression. These findings aligns with prior evidence that elevated glutamine metabolism supports tumor growth and therapy resistance in gliomas [5], indicating that metabolic imaging may still offer complementary insights, particularly when targeting specific metabolic pathways.
Wafae LABRIJI, Amel AISSAOUI, Lotfi CHAARI, Jean-Yves TOURNERET, Elizabeth MOYAL COHEN-JONATHAN, Soleakhena KEN (Toulouse)
15:44 - 15:46
#47761 - PG104 A multicenter classifier of IDH status in gliomas based on in vivo spectroscopy metabolic biomarkers.
PG104 A multicenter classifier of IDH status in gliomas based on in vivo spectroscopy metabolic biomarkers.
Gliomas are the most common primary brain tumors, derived from glial cells and classified according to the 2021 WHO guidelines.
The isocitrate dehydrogenase (IDH) mutation is associated with a better prognosis, and leads to the accumulation of D-2-hydroxyglutarate (2HG), a metabolite measurable by magnetic resonance spectroscopy (MRS) with 100% specificity [2].
Despite its diagnostic value, the clinical detection of 2HG remains limited.
There is a growing need for additional non-invasive metabolic biomarkers to distinguish IDH-mutant from wild-type gliomas without surgery, in order to optimize clinical decision-making.
This study aims to develop an automatic classifier based on multicenter in vivo MRS data to non-invasively differentiate IDH-mutant from IDH-wildtype gliomas.
It focuses on identifying metabolic biomarkers complementary to 2HG, whose clinical detection remains challenging, to support diagnosis according to the 2021 WHO classification.
A multicentric cohort of 186 patients was analyzed with histological classification based on the 2021 WHO guidelines. MR spectroscopy data were acquired on 3T Siemens scanners using single-voxel MEGA-PRESS sequences [2] with water suppression and standardized voxel sizes (>3cm³) to ensure optimal signal quality. Spectra were corrected for frequency and phase shifts and quantified using LCModel [3], with metabolite concentrations normalized to total choline. Quality control included linewidth thresholds for total creatine. Data harmonization was performed using NeuroCombat [4], followed by statistical selection of significant metabolites via Kruskal-Wallis testing with multiple comparison correction. Machine learning classifiers—including PLS-DA, random forests, support vector machines, XGBoost, and an ensemble logistic regression model—were trained on balanced data using SMOTE to address class imbalance (Figure 1). Statistical analysis using the Kruskal-Wallis test identified several metabolites significantly associated with IDH mutation status, including 2HG, alanine, glutamate, glutamine, glutathione, glycine, lipids, GABA, myo-inositol, cystathionine, citrate, betaine, and total creatine (Figure 2). Representative in vivo edited and non-edited MR spectra from IDH-mutant and wild-type gliomas demonstrated clear metabolite distinctions. Edited spectra highlighted the 2HG signal with model fits, while non-edited spectra reveal additional significant metabolites, supporting metabolic profiling linked to molecular status (Figure 3). Multivariate classification models trained on these metabolites achieved high performance in distinguishing IDH-mutant from wild-type gliomas. Notably, the ensemble model outperformed 2HG alone, reaching an AUC of 0.99 compared to 0.78 (non-edited) and 0.85 (edited) for 2HG (Figure 4). The superior performance of multivariate models, particularly the ensemble classifier achieving an AUC of 0.99, highlights the advantage of integrating complementary metabolic biomarkers for more robust and accurate tumor characterization. This suggests that relying solely on 2HG measurements may miss important diagnostic information that can be captured through a wider metabolic signature. These findings highlight the effectiveness of combining advanced magnetic resonance spectroscopy with multivariate machine learning models to noninvasively stratify glioma patients according to molecular status. This integrated approach provides a more comprehensive metabolic profile of tumor heterogeneity, paving the way for faster, more precise clinical decision-making and improved personalized treatment planning.
Capucine CADIN (Paris), Gerd MELKUS, François-Xavier LEJEUNE, Dinesh DEELCHAND, Stéphane LEHÉRICY, Malgorzata MARJANSKA, Thanh BINH NGUYEN, Francesca BRANZOLI
15:46 - 15:48
#46286 - PG105 Metabolic CEST MR imaging of glucosamine uptake in brain tumors.
PG105 Metabolic CEST MR imaging of glucosamine uptake in brain tumors.
Abnormal metabolism is a known phenomenon in many brain diseases, particularly brain cancer, that is characterized by a rapid and accelerated metabolism of tumor cells. Thus, imaging techniques capable of detecting metabolic alterations are critical for detecting brain cancer at an early stage. Chemical exchange saturation transfer (CEST) MRI is an increasingly investigated imaging technique that enables non-invasive metabolic activity measurement in vivo. Glucose (Glc) uptake is a sensitive biomarker for brain cancers in their early stages1, hence, monitoring Glc uptake and metabolism in the brain may be highly useful in the clinic, as extensively shown in previous glucose2 and its analog studies3.
We recently demonstrated the ability of glucosamine (2-amino-2-deoxy-D-glucose, GlcN) CEST MRI to detect breast cancer in mice4,5, and humans6 as well as to detect the uptake of GlcN in the healthy mouse brain7. Here we hypothesize that GlcN uptake in brain tumors can be detected using the CEST MRI technique. In this study, we imaged in vivo tumor-bearing mice brains before and after GlcN injection and analyzed the data using the magnetic transfer ratio asymmetry (MTRasym) metric. We demonstrate the feasibility of using GlcN as a CEST agent to detect brain cancer in a mouse model.
Animal Preparation: All animal experiments and procedures followed the principles of the Israel National Research Council (NRC) and were approved by the Tel Aviv University Institutional Animal Care and Use Committee (IACUC) (TAU-MD-IL-2405-129-5) . Male C57BL/6J mice (6-week-old, n=6) were purchased from Harlan (Israel). GL261 cells (1X105) were injected for each mouse under anesthesia using stereotactic control. Mice were studied by MRI at 10-17 days after implanting the cells.
MRI Acquisition: MRI experiments were performed on a preclinical 7T scanner (Bruker, Germany) with a volume transmit-receive coil (internal diameter 38 mm, Bruker Biospin, Germany), equipped with a Bruker Paravision software (PV6) (Bruker Biospin, Germany). During the MRI scan, mice brains were scanned using a quadrature coil, they were anesthetized using isoflurane (0.8%) and midazolam (2 mg/kg, SC)8 in a mixture of O2 and air gases and kept warm with a heated bed. Mice were fasted for at least 3 h (with water access) prior to the experiments. An anatomical T2 scan was performed to locate the slice of the tumor. CEST imaging was conducted before and after intravenous (IV) injection of GlcN sulfate (2.0 g/kg, IV, dissolved in saline, via venflon, 27G needle with cone filled with heparin, ~0.3l). The field of view (FOV) was 19 × 19 mm, a matrix (MTX) of 64 × 64 pixels, and the slice thickness of 1 mm. Z-spectra were obtained using a CEST-EPI protocol 10, employing a saturation pulse power of 2.5 μT, saturation pulse time (Tsat) = 2s, TE/TR = 20/8000 ms, and saturation pulse frequency offsets of 7 to −7 ppm with 0.25 ppm increments (acquisition time = 7:45 min:s). For the static magnetic field calculation, a B0 map was acquired. Also, the WASSR9 method was used (fat suppressed turned on, NA=1).
Data Processing: CEST signals were analyzed in the tumor and the contralateral region of interests (ROIs) using the magnetization transfer ratio with asymmetric analysis (MTRasym) metric and the area under the curve (AUC) method (between 1 to 2.2 ppm, representing hydroxyls contribution). All data were processed in Python. Figure 1 shows the AUC maps (1-2.2 ppm) obtained from three representative tumor-bearing mice at baseline, 8 minutes, and 16 minutes after GlcN injection. An increased MTRasym signal (Figs.2,3) was obtained in the tumor’s ROI compared to the contralateral tissue and baseline conditions. The GlcN injection consistently elevated the AUC signal at the tumor for all mice and time points. The contralateral ROI signal was elevated in some cases following the injection, as previously reported7; however, the signal increase was significantly higher at the tumor (Fig. 4, P = 0.037 after 8 minutes, and P = 0.014 after 16 minutes). Hydroxyls, NOE and Amides contribute to the AUC signal and may decrease each other’s effect. We suggest separating the contribution of these solutes using MRF in future research. This is the first CEST MRI study examining GlcN uptake and efficacy in the pathological brain. The technique may open new avenues for metabolic brain imaging using a safe and bio-compatible food supplement, potentially aiding in early brain tumor detection.
Yaniv YOSHA (Tel Aviv, Israel)
15:48 - 15:50
#47367 - PG106 Quantitative analysis of segmented whole-brain MRSI in epilepsy: an initial report.
PG106 Quantitative analysis of segmented whole-brain MRSI in epilepsy: an initial report.
Epilepsy is a heterogeneous brain disorder that causes recurrent seizures. Over 65 million patients around the world are diagnosed with epilepsy, where 30% of them cannot be treated sufficiently with anti-seizure-medication [1,2]. Currently, the most efficient treatment for these patients is to undergo surgery and resect the Epileptic Zone (EZ). The best-known biomarker for EZ is the Seizure Onset Zone (SOZ), which triggers the seizures. Although various modalities are used in presurgical evaluations, 30% of patients are not seizure-free after surgery.
Ultra-high-field (UHF) magnetic resonance imaging (MRI) at 7T has a higher signal-to-noise ratio which leads to better structural irregularities detection compared to lower-field scanners [3].
Despite using a 7T scanner no structural irregularities can be detected for a considerable number of patients who are called magnetic resonance negative (MRN) [4, 5].
Magnetic Resonance Spectroscopic Imaging (MRSI) images neurochemical concentrations [6]. Detecting neurochemical changes holds significant potential for identifying abnormalities in patients with MRN epilepsy. Previous studies have utilized single-voxel magnetic resonance spectroscopy, which cannot cover the entire MRN brain [7]. We recently developed a new method for fast and high-resolution whole-brain 3D MRSI at 7T and applied it to an initial cohort of patients in a qualitative analysis [8, 9].
In this study, we aimed to analyze median concentration estimates in parcellated regions of the brain as a step toward clinical evaluation of 7T MRSI for epilepsy.
Our retrospective dataset included 7T MRI and MRSI scans from 10 epilepsy patients, 8 of them MRN (Table 1). We use a 7T scanner (Magnetom Plus, Siemens, Erlangen, Germany) with a 32-channel head coil, local IRB approval, and informed consent. Our protocol contained 3D CRT-FID-MRSI (TR=450ms, acquisition delay=1.3ms, FOV=220×220×133mm³, resolution=3.4×3.4×3.4 mm³, scan time=15 min). The MRSI data was post-processed and quantified using MATLAB [10] and LCModel [11], with a focus on glutamine (Gln), glutamate (Glu), N-acetyl aspartate (NAA), creatine + phosphocreatine (tCr), and myo-inositol (Ins) [9]. White matter/ gray matter segmentation and literature values for proton density were used to calculate concentration estimates [12]. Figure 1 illustrates data processing and evaluation. In the first step, we used “cortex parcellation” of SynthSeg [13] to segment the T1-weighted (T1w) images with 0.75×0.75×0.75 mm³ resolution. We then applied the segmentation output to the neurochemical maps and calculated median concentration estimates in the right and left hemispheres for the frontal, parietal, temporal, and occipital lobes. As a marker of abnormality, we calculated a relative difference between the two hemispheres right hemisphere - left hemisphereleft hemisphere100 which we compared with the clinical findings (Table 1). Table 1 shows regional medians and their evaluation for tCr. The inter-hemisphere asymmetries in each lobe were calculated by comparing differences in each lobe. The highest difference values in each column (among all patients) are marked with a red rectangle. For tCr in Table 1, we have 4 asymmetries found in the SOZ region (green boxes/Y) and 2 were found out of SOZ (red boxes/N). This is summarized for all metabolites and the results are summarized in Table 2. Table 3 compares the median of CE’s relative differences among all patients. The highest difference percentages (asymmetries) in the temporal lobe in 4/5 metabolites fit well in the 7/10 patients with temporal lobe epilepsy in the dataset. The highest relative difference among all metabolites belongs to Gln followed by Glu. We successfully determined median concentration estimates in brain ROIs. The metabolic inter-hemisphere asymmetry aligned well with the SOZ in 7 out of 10 patients, This finding needs further investigation for more robust clinical use. Overall, we found the highest asymmetries for Gln, with a maximum in the temporal lobe which was the most common SOZ in our cohort. Our findings are consistent with previous literature that used MRS to lateralize the SOZ [7].
This study was limited by a small cohort and coarse GM ROIs. In the future, we will evaluate more patients and metabolites in a fine-grained parcellation of the brain. Most importantly, including healthy controls matched for age and sex will enhance our analysis This study gives an overview of the quantitative analysis of the MRSI maps segmented in main lobes. While there are many more capacities for further quantitative analysis, this report represents an initial successful output of the quantitative analysis of MRSIs of epileptic patients.
Haniye SHAYESTE (wien, Austria), Stefanie CHAMBERS, Philipp LAZAN, Matthias TOMSCHIK, Jonathan WAIS, Gregor KASPRIAN, Lukas HAIDER, Leo HOFER, Christoph BAUMGARTNER, Johannes KOREN, Martha FEUCHT, Florian MAYER, Silvia BONELLI, Christian DORFER, Ekaterina PATARAIA, Wolfgang BOGNER, Siegfried TRATTNIG, Karl RÖSSLER, Gilbert HANGEL
15:50 - 15:52
#46459 - PG107 Assessment of inter- and intra epileptic network functional connectivity changes in focal epilepsy patients using 7T MRI.
PG107 Assessment of inter- and intra epileptic network functional connectivity changes in focal epilepsy patients using 7T MRI.
Focal epilepsy is a chronic neurological disorder characterized by recurrent, spontaneous seizures originating from abnormal neuronal activity. Seizures propagate from the epileptogenic zone (EZ) network to connected regions within the propagation zone (PZ) network [1], potentially leading to long-term alterations in brain function. Stereotactic electroencephalography (SEEG) recordings in drug-resistant focal epilepsy (DRFE) patients have shown increased functional connectivity among EZ and PZ networks between seizures [2], while MRI studies have reported reduced functional connectivity [3]. This apparent discrepancy may reflect methodological differences: prior MRI studies primarily assessed intra-subject connectivity changes, without considering spatially-specific alterations relative to healthy controls. It remains unclear how the MRI-detected connectivity decreases are in comparison to healthy controls and/or confined to non-epileptogenic regions. To address this, we assessed resting-state functional MRI (rs-fMRI) connectivity changes in DRFE patients relative to healthy controls, focusing on SEEG-defined networks.
We retrospectively included 23 DRFE patients from the ‘Multimodal Whole Brain Imaging in Epilepsy’ (MOBILE) dataset by the European Human Brain Project [4], and 42 matched healthy controls. Inclusion criteria included the evaluation with SEEG and availability of high quality 7 Tesla (7T) anatomical and functional MRI data with no signs of brain surgery or major brain malformations. The MOBILE study received approval from the local ethics committee, and all participants provided written informed consent.
MRI data acquisition included B1+ and 0.6 mm3 T1 mapping, and 1.6 mm3 rs-fMRI, using either a whole-body 7T MAGNETOM or MAGNETOM TERRA scanner (Siemens Healthineers, Erlangen, Germany) with a 1H 1Tx/32Rx (Nova Medical, Wilmington, USA) head coil. SEEG recordings were performed as part of the clinical care following French clinical guidelines [5].T1 images were B1+ corrected T1, skull-stripped, and segmented using FreeSurfer [6]. Cortical and subcortical regions were defined using the Virtual Epileptic Patient (VEP) atlas and served as regions-of-interest for connectivity analyses [7]. The rs-fMRI data were preprocessed using fMRIprep (v20.2.1), followed by nuisance regression using the strategy described in [8]. SEEG-derived epileptogenicity index (EI) classified regions as EZ (EI ≥ 0.4), PZ (0.1 ≤ EI < 0.4), non explored (NI) (EI < 0.1), or NE (not explored) [1].Functional connectivity was computed as Pearson correlation between VEP region pairs. Deviations from control connectivity were z-scored per patient, adjusted for age, sex, and scanner. Median z-scores characterized each "connectivity type". Functional connectivity differences between patients and controls were subtle across all connection types (Fig. 2). Nonetheless, z-score analysis revealed significant variability across connectivity types (F9,111.5=8.39, p<0.0001), with EZ–EZ, EZ-PZ, and PZ–PZ connections showing connectivity values closer to those of healthy controls than the rest (F1,150.3=54.51, p<0.0001), albeit with increased inter-subject variability (Fig. 2). On average functional connectivity between EZ regions was slightly decreased as compared to controls (Z-score= -0.24, Fig. 2). Notably, connectivity was consistently higher within and between EZ and PZ regions when the thalamus was part of either their networks (F1,27.5=13.84, p=0.0009, Fig. 3A). Additionally, mean SEEG-derived epileptogenicity values were significantly correlated with functional connectivity z-scores (r=-0.31, p<0.0001, Fig. 3B) This study provides evidence that functional connectivity alterations in DRFE patients are spatially heterogeneous and modulated by epileptogenicity. Contrary to prior group-level MRI studies suggesting widespread hypoconnectivity [3], we found that functional connectivity with(in) NI and NE zones was slighty higher than in controls. On the other hand functional connectivity between SEEG-defined EZ and PZ was relatively preserved, although variable, or slighty decreased for EZ, in line with a previous structural connectivity study [9]. This discrepancy was especially pronounced when the thalamus was not participating in the EZ or PZ, underscoring the thalamus’s potential role in seizure dynamics, as supported by both intracerebral recordings and recent 7T MRI studies showing thalamic structural and microstructural alterations [10]. This study shows that static rs-fMRI connectivity in SEEG-defined networks of DRFE patients is spatially heterogeneous and modulated by epileptogenicity.These results underscore the need for individualized, multimodal network specific analyses to better understand and eventually target pathological connectivity in epilepsy. Future work will assess dynamic connectivity and its relation to epileptogenicity.
Tatiana HOROWITZ, Julia MAKHALOVA SCHOLLY, Hugo DARY, Samuel MEDINA VILLALON, Jean-Philippe RANJEVA, Fabrice BARTOLOMEI, Maxime GUYE, Roy HAAST, Roy HAAST (Marseille)
15:52 - 15:54
#46971 - PG108 Morphometric changes of hypothalamic nuclei in drug-resistant focal epilepsy observed using 7T MRI - Relationships to anxiety and post traumatic stress syndrome.
PG108 Morphometric changes of hypothalamic nuclei in drug-resistant focal epilepsy observed using 7T MRI - Relationships to anxiety and post traumatic stress syndrome.
Multiple studies have emphasized the involvement of subcortical structures such as the thalamus and amygdala in epileptic networks. Nonetheless, the role of the hypothalamus (HT) remains little explored, despite its involvement in regulating stress, sleep, and neuroinflammatory processes. This study aims to use ultra-high field (7T) MRI to characterize the volumes of individual hypothalamic nuclei in patients with drug-resistant focal epilepsy (DRFE), investigating possible structural alterations associated with this disease and with its psychiatric comorbidities such as anxiety and post-traumatic stress disorder (PTSD).
A total of 79 DRFE patients and 66 healthy controls underwent 7T MRI (TERRA Siemens, Erlangen, Germany) using a 1Tx/32RX head coil (NOVA Medical). All subjects gave their informed consent in accordance with the declaration of Helsinki. Anxiety and PTSD symptoms were assessed using validated psychiatric screening scales (GAD-7, PCL-5, PTSD-E).
The anatomical MRI protocol included B0 shimming, a B1+ map and a high-resolution 3D T1-weighted MP2RAGE sequences (TR=5000 ms, TE=3 ms, TI1=900 ms, TI2=2750 ms, 256 partitions, isotropic voxel size=0.6mm, TA=10 min). Images were post-processed according to [1] to compute UNIDEN images corrected for B1+ inhomogeneities [2]. After ANTS coregistration of the Neudorfer atlas template [3] to each subject’s corrected UNIDEN image, volumes of individual HT nuclei were extracted and z-scored relative to the mean and standard deviation across the healthy controls.
Non-parametric Mann Whitney U-tests (Bonferroni corrected) were performed to assess z-score differences between patients and controls for each nucleus. Paired Wilcoxon tests were also conducted in patients to compared z-scores values between homologous HT nuclei ipsilateral and contralateral to the seizure onset side. Finally, the non-linear partial least square model (NIPALS) was used to study the association of HT volumes with anxiety and PTSD scores. Compared to controls, patients showed significant volume increases (p<0.0038) for the ipsilateral dorsomedial (DM) and posterior (PHN) HT nuclei, as well as the contralateral ventromedian (VM), PHN, lateral (LH), anterior (AHA) and tuberomammillary (TM) nuclei (Figure 1). No volume differences were observed between corresponding ipsilateral and contralateral HT nuclei in DRFE patients.
Partial least squares analyses revealed strong associations between anxiety severity (GAD-7 scores) and increased volumes of the contralateral VM, AHA, median preoptic (MnPO) and supraoptic (SON) nuclei (Figure 2). Similarly, PTSD symptom severity (PTSD-E scores) was strongly associated with increased volumes of the contralateral MnPO, contralateral SON and ipsilateral LH nuclei (Figure 3). We observed significant volume increases of several HT nuclei previously reported to be implicated in seizure susceptibility and comorbid neuropsychiatric conditions. The DM and PHN have been linked to seizure generation and panic disorder [4], the VM [5], PHN [4], LH [6] and AHA [7][8] are involved in stress regulation and anxiety, while the TM is implicated in sleep-pain coregulation [9].
Although the volumes of MnPO and SON did not differ significantly from controls, they contributed to the variance in anxiety and PTSD symptom severity. This aligns with their involvement in the neural networks of depression [10] and stress regulation [11] through the HPA axis [12]. Ultra-high field 7T MRI enables the detection of structural alterations in specific HT nuclei in DRFE patients. Our observations highlight the hypothalamus as a potential contributor to the complex network underlying epilepsy and its psychiatric comorbidities.
Manel KROUMA (Marseille), Julia MAKHALOVA, Hugo DARY, Roy HAAST, Lauriane PINI, Shirley CORELLA, Francesca PIZZO, Stanislas LAGARDE, Maxime GUYE, Fabrice BARTOLOMEI, Jean-Philippe RANJEVA
15:54 - 15:56
#47318 - PG109 Characterization of drug-resistance in patients with temporal lobe epilepsy: insights from single-subject T1 mapping.
PG109 Characterization of drug-resistance in patients with temporal lobe epilepsy: insights from single-subject T1 mapping.
Recently, normative atlases for T1 relaxation times have been proposed for individualized quantification of brain tissue abnormalities[1]. In mesial temporal lobe epilepsy (MTLE), the most common form of epilepsy in adulthood, a large body of evidence suggests that MTLE-related pathophysiological changes may extend beyond the temporal lobe, through cortico-cortical and cortico-subcortical networks[2].
In this study, we described the distribution of quantitative T1 alterations across white matter (WM) and subcortical regions in patients with MTLE to identify potential quantitative correlates of drug-resistance, which might be useful to improve patient treatment using individually tailored options.
Thirty-eight patients (23 females; age: 38.97 ± 14.24 years) with MTLE underwent 3T MRI (Biograph mMR, Siemens Healthineers, Forchheim, Germany) using a 16-channel PET-transparent head/neck coil. The protocol included an MP2RAGE sequence[3] with the following parameters: TI1/TI2/TE/TR=700/2500/2.96/5000 ms; flip angles 4/5 deg; 1 x 1 x 1 mm3; GRAPPAx3 [4] . At the time of the MRI exam, to evaluate the effect of treatment response we identified 9 out of 38 patients drug-naive that started therapy shortly after diagnosis and came back for a clinical follow-up, resulting in 7 drug-responsive and 2 drug-resistant patients observed before treatment. Baseline EEG recordings showed maximal epileptiform discharges (side of EEG) over left temporal regions in 20 out of 38 and over right temporal regions in 9 out of 38 patients. Single-subject comparisons of voxel-wise quantitative parameters against age-/sex matched healthy reference values were obtained following Piredda et al[1]. Briefly, individual T1 maps were co-registered to a normative atlas, established using linear regression models, to consider each patient’s age and sex when calculating deviations from the healthy population[1,4]. For each individual patient, such deviations were evaluated as voxel-wise z-scores in WM and subcortical tissues.
Individual z-score maps in the atlas space were thresholded and binarized to maintain only voxels with more than 2 standard deviations from the corresponding normative population value (i.e., |z| > 2). Using FSL utils[5], we obtained 4D images of individual z-score maps.
Group-wise analyses were performed on percentage maps for drug-naive MTLE patients at MRI time and drug-responsive at clinical follow-up while single-subject assessments were conducted in patients drug-naive at MRI time and drug-resistant at clinical follow-up. In all comparisons no negative deviations were observed (z<2). Comparing deviated voxel count in patients drug-naive at MRI examination, we observed that MTLE patients drug responsive at clinical follow-up (n=7) were relatively free from alterations (Figure 1). Individual T1 z-score maps of two patients who were drug-naïve at the time of MRI, started therapy shortly after diagnosis and came back for a clinical follow-up after 9 months due to drug-resistance to ASMs, in both cases (Figures 2 and 3) z-score analysis confirmed the presence of alterations distributed in the hippocampus and in temporal lobe WM, as well as in subcortical structures namely striatum and thalamus. It is well known that MTLE affects temporal regions of the brain, and that HS has long been considered a hallmark of drug-resistant epilepsy. These results support and strengthen this piece of knowledge, by providing a quantitative assessment of the frequency and extent of detected abnormalities in these regions, especially in patients that don’t respond to ASMs, in which seizure recurrence may affect brain structural integrity. Moreover, striatal involvement in epilepsy is less investigated, but still associated with focal impaired awareness seizure, frequently observed in MTLE. Indeed, quantitative assessment of brain tissue damage due to seizure recurrence in drug-resistant patients represents a promising measure to improve patient care and monitoring over time, potentially helping clinicians to define better individually tailored treatment strategies, as well as to identify early severe drug-resistant patients as candidates for epilepsy surgery.
Maria Celeste BONACCI (Catanzaro, Italy), Veronica RAVANO, Ilaria SAMMARRA, Gian Franco PIREDDA, Anais BURRUS, Domenico ZACÀ, Bénédicte MARÉCHAL, Tom HILBERT, Tobias KOBER, Antonio GAMBARDELLA, Maria Eugenia CALIGIURI
15:56 - 15:58
#47827 - PG110 Iron-related susceptibility changes in brainstem arousal centres in adult narcolepsy type 1: insights from quantitative susceptibility mapping.
PG110 Iron-related susceptibility changes in brainstem arousal centres in adult narcolepsy type 1: insights from quantitative susceptibility mapping.
Narcolepsy type 1 (NT1) is a rare sleep disorder characterized by excessive daytime sleepiness and cataplexy, probably linked to the loss of orexin-producing neurons in the hypothalamus.
Advanced neuroimaging techniques such as quantitative susceptibility-mapping (QSM) can detect magnetic susceptibility alterations, often reflecting iron accumulation; in our preliminary study [1], we showed hypothalamic increased susceptibility that we linked to possible localized neurodegeneration phenomena.
Hypothalamus is connected to various brain regions, including brainstem where its nuclei play essential roles in arousal and sleep-wake regulation.
This study aimed to characterize QSM-based susceptibility changes in brainstem nuclei of adult NT1 patients, potentially indicating broader structural involvement in the disease.
63 NT1 patients (40M, 34±12 years) and 54 healthy controls (26M, 41±11 years) underwent a 3T MRI protocol including T1-weighted (3D-MPRAGE, TR/TE=2300/2.98 ms, 1×1×1 mm³) and QSM (3D multi-echo GRE T2*-weighted, nTEs = 5, TR/TE/ΔTE = 53/9.42/9.42 ms, 0.5×0.5×1.5 mm³). Magnetic susceptibility (χ) maps were reconstructed from the raw QSM [2]. A probabilistic atlas of the brainstem [3] was registered to the individual T1-weighted images to segment the following subregions: dorsal raphe (DR), locus coeruleus (LC), laterodorsal tegmental nucleus (LDTg), median raphe (MnR), mesencephalic reticular formation (mRt), periaqueductal grey (PAG), parabrachial complex (PBC), pontis oralis (PnO), pedunculotegmental nucleus (PTg), and ventral tegmental area (VTA). QSM images were co-registered to T1 space to extract χ values [4] from the segmented subregions (Figure 1). Raw data were age-corrected using linear regression based on the control group. Normality was assessed using the Shapiro-Wilk test, and statistical comparisons were performed using ANOVA or the Mann-Whitney U test. NT1 patients showed significantly higher χ values compared to controls in several brainstem subregions, including the LC (p = 0.01), LDTg (p = 0.02), MnR (p = 0.0026), PBC (p = 0.01), and PnO (p = 0.004), after correction for multiple comparisons (Figure 2). No significant group differences were found in the other segmented regions. This study provides novel evidence of altered magnetic susceptibility in key brainstem arousal nuclei in adult NT1 patients, as revealed by quantitative susceptibility mapping (QSM). Significantly increased χ values were detected in the locus coeruleus (LC), laterodorsal tegmental nucleus (LDTg), median raphe (MnR), parabrachial complex (PBC), and pontis oralis (PnO, i.e. pontine reticular formation). These regions are integral components of the ascending arousal system and some of them are known to receive direct projections from orexin-producing neurons, which are selectively lost in NT1.
The observed QSM alterations likely reflect increased iron deposition, a well-established surrogate marker for neurodegeneration [5]. Iron accumulation in the brain can result from microglial activation, oxidative stress, or impaired clearance mechanisms, and is commonly associated with neuronal loss or dysfunction. The spatial distribution of susceptibility changes, notably across multiple monoaminergic and cholinergic nuclei, suggests that NT1 may involve more widespread pathological processes than previously appreciated, extending beyond the hypothalamus to downstream arousal centres in the brainstem.
Moreover, several of the affected nuclei—such as the LC and MnR—are involved not only in sleep-wake regulation but also in mood, autonomic function, and attention, which could be related to some of the non-sleep-related symptoms frequently reported in NT1 patients. The present findings therefore support the hypothesis that NT1 is not solely a hypothalamic disorder but may be part of a broader neurodegenerative process involving key subcortical circuits. Increased magnetic susceptibility in multiple brainstem arousal nuclei in NT1 patients, as measured by QSM, provides in vivo evidence of a possible diffuse neurodegenerative process [5]. These findings extend the neuroanatomical scope of NT1 beyond the hypothalamus and highlight the potential utility of QSM as a valuable tool for detecting subtle subcortical changes associated with sleep-wake disorders. Future longitudinal studies are warranted to clarify the temporal evolution and clinical relevance of these alterations.
Lorenzo MOTTA (Bologna, Italy), Greta VENTURI, Francesco BISCARINI, Fabio PIZZA, Raffaele LODI, Giuseppe PLAZZI, Caterina TONON
15:58 - 16:00
#47731 - PG111 Lifespan iron and neuromelanin accumulation in dopaminergic neurons.
PG111 Lifespan iron and neuromelanin accumulation in dopaminergic neurons.
Dopaminergic neurons in the substantia nigra (SN) are among the cells with the highest iron concentrations in the brain. They require sufficient iron for neurotransmitter synthesis, but are affected by iron-induced oxidative stress when iron levels become high. To prevent this, iron is stored in neuromelanin, a biopolymer composed of proteins, lipids and iron-chelating chromophores. However, neuromelanin may become toxic when iron-overloaded with age, increasing the risk of Parkinson’s disease[1]. A non-invasive method to monitor the iron and neuromelanin balance is needed to detect Parkinson’s disease at an early stage and to understand its underlying mechanisms.
Quantitative maps of the effective transverse relaxation rate R2* have been shown to be sensitive to the tissue iron concentration. Recently, a biophysical model linked R2* to the iron load and neuronal density of dopaminergic neurons in SN[2]. Moreover, neuromelanin-sensitive T1 and MT contrasts are promising biomarkers of neuromelanin in SN showing plausible results in post mortem tissue. However, these methods have only been validated in post mortem brains of elderly donors and the mechanisms of iron- and neuromelanin-driven contrast in younger brains remain to be investigated. The relationship between iron and neuromelanin accumulation in dopaminergic neurons across the lifespan remains unknown. This is challenging, since post mortem tissue from young humans is rare and common lab animals are not useful due to short lifespans and low iron levels.
Combining ultra-high resolution quantitative relaxometry, iron quantification by synchrotron X-ray fluorescence (XRF) and optical spectroscopy, we compared the lifespan trajectory of R2*, iron and neuromelanin in ethically collected post mortem chimpanzee brains[3]*.
Quantitative R2* maps of 22 post mortem chimpanzee brains aged from 1 month to 52 years were acquired at 7T using multiparametric mapping[4] with an isotropic resolution of 300µm. Median R2* in the SN were used to infer tissue iron concentrations and compared against human data[5]. Cellular iron concentrations in the automatically segmented neuromelanin clusters within dopaminergic neurons were obtained by XRF at beamline P06, PETRA III (DESY, Hamburg) and were compared against iron concentrations in human dopaminergic neurons. The lifespan trajectories of R2* and iron concentrations were modeled by an exponential saturation[6] using a Bayesian approach. Optical spectroscopy with cellular resolution was used to estimate the neuromelanin concentration within the neurons and its iron binding capacity. A chemical equilibrium model considering iron binding in two binding sites within neuromelanin was used to estimate the magnetic susceptibility of neuromelanin-bound iron and lifespan changes in free cytosol iron reflecting iron toxicity. Both the R2* in the SN and the cellular iron concentration of dopaminergic neurons increased with age, but the time constants of these increases differed. The cellular neuromelanin-iron accumulation measured by XRF in a large group of neurons was well described by an exponential saturation with a time constant of about 27±5 years (posterior max ±50% highest density interval, Fig.2). In contrast, the averaged tissue iron concentration in the SN increased much slower (time constant of 39±10 years).
In adult age, the R2* in the SN of chimpanzees (mean 65.7 1/s, age >20y) corresponded to those in humans[5] (69.2 1/s, Fig.1), indicating similar overall tissue iron concentrations in both species. The cellular iron concentrations in young chimpanzees were comparable to those in young humans[7](Fig.2).
The neuromelanin concentration within dopaminergic neurons increased with age, showing a linear relationship with the iron concentrations in the same neurons, with similar slopes across ages and species([2]Fig.3). We estimated the maximal bound iron[8], the neuromelanin iron saturation and the concentrations of potentially neurotoxic free iron by modeling the iron binding in neuromelanin and measuring cellular iron and neuromelanin concentrations. Different time trajectories of cellular iron and R2* point to different underlying physiological processes for iron accumulation in dopaminergic neurons and surrounding tissue.
Assuming different susceptibilities of iron bound to different sites, a change in neuromelanin saturation has implications on the susceptibility of neuromelanin across the lifespan. Using XRF and qMRI, we characterized the age-related iron and neuromelanin accumulation in dopaminergic neurons in the SN of our closest relatives, chimpanzees. We demonstrated that the chimpanzee is a suitable animal for studying the human SN across the lifespan. Iron accumulated faster in dopaminergic neurons than in the entire SN, as measured by R2* and closely followed the accumulation of neuromelanin across lifespan. This data will inform biophysical modeling to understand the contrast mechanism in SN across the human lifespan.
Felix BÜTTNER (Leipzig, Germany), Tilo REINERT, Carsten JÄGER, Malte BRAMMERLOH, Markus MORAWSKI, Ilona LIPP, Gerald FALKENBERG, Dennis BRÜCKNER, Catherine CROCKFORD, Roman WITTIG, Nikolaus WEISKOPF, Evgeniya KIRILINA
16:00 - 16:02
#47935 - PG112 Voxel-wise Estimation of Optimal Intrinsic Diffusivity in NODDI Reveals Increased Values in Alzheimer’s Disease White Matter.
PG112 Voxel-wise Estimation of Optimal Intrinsic Diffusivity in NODDI Reveals Increased Values in Alzheimer’s Disease White Matter.
In the context of Multiband Diffusion Tensor Imaging (DTI), the Neurite Orientation Dispersion and Density Imaging (NODDI) model provides a direct estimation of brain microstructure by modeling the tissue as a multi-compartmental system, accounting for free, hindered and restricted water diffusion[1]. Expressing the diffusion MRI (dMRI) signal as a sum of contributions from each of the three compartments modeled, NODDI provides specific metrics (i.e. Neurite Density Index (NDI) and Orientation Dispersion Index (ODI)) which are able to quantify the effects of density and orientation distribution of dendrites and axons. The NODDI model has been shown to enable an improved assessment of white matter (WM) microstructure, offering greater specificity than the more common DTI metrics of Fractional Anisotropy (FA) and Mean Diffusivity (MD). For this reason, this model has been increasingly used to assess microstructural alterations in WM in a variety of diseases, with metrics often resulting more sensitive to changes and thus better for diagnostic purposes[2,3]. A critical parameter in the NODDI model is the axial intrinsic diffusivity (d||) of the neurite compartment, which is typically fixed at 1.7×10⁻⁹ m²/s. Although this default value is optimized for adult WM, recent studies suggest that it may be suboptimal in infant brains and in gray matter of adult brains[4]. We hypothesized that d|| might also deviate in Alzheimer’s Disease (AD) WM, with optimal values being higher than the assumed 1.7×10⁻⁹ m²/s due to neurodegeneration. The aim of this study is to explore whether the optimal intrinsic diffusivity value in WM of AD patients might be significantly different compared to controls.
We analyzed multiband dMRI data from 145 subjects in the ADNI dataset[5]: 26 AD patients, 60 Mild Cognitive Impairment (MCI) patients, and 59 Cognitively Normal (CN) controls. Following the optimization process proposed by Guerrero et. al.[4], we used the Microstructure Diffusion Toolbox (MDT)[6] to perform NODDI model fitting for 15 different values of d|| (equally spaced from 0.8×10⁻⁹ to 2.5×10⁻⁹ m²/s). For each model fit, the MDT toolbox pipeline output voxel-wise log-likelihood maps, which were then used to derive a “best- d||” map indicating the d|| value maximizing the model likelihood at each voxel. Diffusion data preprocessing was conducted using MRtrix3[7] and FSL[8] (denoising, Gibbs ringing removal, bias correction, eddy-current and motion correction). FA maps were extracted and used to generate a white matter skeleton via Tract-Based Spatial Statistics (TBSS)[9]. The “best-d||” maps were smoothed (σ=1.5) to flatten out unwanted outliers and projected onto the mean FA skeleton. TBSS analysis with threshold-free cluster enhancement (TFCE) was performed for the contrasts AD>CN, AD>MCI, and MCI>CN, in order to evaluate whether the optimal intrinsic diffusivity value would be higher in any white matter tracts in patients compared to controls. Significant clusters (p<0.05) were observed for AD>CN and AD>MCI, particularly in the corpus callosum and bilateral cingulum, where AD patients showed higher optimal intrinsic diffusivity values compared to CN and MCI. No significant differences were found for MCI>CN at p<0.05. This suggests increased optimal d|| in regional WM tracts of AD patients, reflecting altered WM microstructural integrity that is already well documented in the literature[3]. These findings indicate that the fixed intrinsic diffusivity of 1.7×10⁻⁹ m²/s used in standard NODDI fitting may not be optimal in the context of AD-related WM degeneration. The elevated optimal d|| observed in AD patients suggests that intrinsic diffusivity itself may carry disease-relevant information, and that it might be a good marker to better distinguish between MCI and AD patients. This warrants further investigation to assess whether optimizing d|| during model fitting of AD patients would provide increased differentiation especially between MCI and AD patients in NODDI metrics. A possible study would be to compare TBSS results of standard NDI and ODI metrics (obtained with d|| =1.7×10⁻⁹ m²/s) to the “corrected” NDI and ODI (with values drawn from the “best-d||” model per voxel). Increased statistical differences between AD, MCI and CN would confirm the advantage of optimizing the intrinsic diffusivity parameter in NODDI modelling of AD. Voxel-wise estimation of the optimal intrinsic diffusivity parameter in the NODDI model reveals significantly elevated values in regional white matter of AD patients. These findings suggest that the model parameter previously considered fixed may offer additional sensitivity to pathological change and should be revisited in the context of neurodegenerative disease.
Tommaso SCACCO (Roma, Italy), Luca PASQUINI
16:02 - 16:04
#47596 - PG113 Changes of Graph-Theory metrics derived from Structural and Functional Brain Network In depressed patients following Electroconvulsive Therapy.
PG113 Changes of Graph-Theory metrics derived from Structural and Functional Brain Network In depressed patients following Electroconvulsive Therapy.
Electroconvulsive therapy (ECT) is the most effective treatment for pharmacoresistant depression, which affects about one-third of patients [1–3]. The objective of this study was to analyze global metrics of brain network graphs obtained from diffusion MRI and resting state functional MRI before and after ECT to identify biomarkers for monitoring the effects of the therapeutic intervention and predicting its effectiveness.
This prospective longitudinal monocentric study (PI: M. Plaze, GHU Paris, France) involves 43 patients (26 women, 54.1 ± 18.1 years old) suffering from major depressive disorder (DSM-5) treated with ECT and 24 healthy participants (14 women, 49 ± 21 years old). The patients were evaluated at three time points: before ECT (V1), after 5 ECT sessions (V2), and 14 days after the end of the treatment (V3). The evaluations included the MADRS clinical score and brain MRI scans using a GE 3T Discovery MR750 scanner with 3D T1 Fast SPGR anatomical sequences with inversion recovery (TE/TR/TI = 3.2/8.2/400 ms, flip angle = 11°), echo-planar diffusion-weighted multiple-shell sequences (b=200/1500/2500 s/mm², 30/45/60 directions) with isotropic resolution of 2 mm and resting state fMRI with gradient-echo single shot EPI sequence (TR/TE = 2000/23 ms), isotropic resolution of 3mm. A reverse phase-encoding direction sequence (b = 2500 s/mm², 6 directions) was acquired to correct for susceptibility-induced distortions. Evaluations for healthy participants only included one brain scan at inclusion (V1). Diffusion index maps (FA, ADC, NDI, etc.) were calculated using both DTI and NODDI models, along with fiber tractography, using CSD orientation distributions with Mrtrix3 [4–7]. Parcellation of T1 images was performed using Freesurfer to extract 164 cortical and subcortical regions based on the Destrieux brain atlas [8,9]. Brain network graphs consist of nodes representing segmented regions of interest. For structural networks, edges represent white matter bundles reconstructed from tractography. For functional networks, edges represent z-transformed Pearson total correlation [10]. Global network metrics (efficiency, robustness) were calculated using R. The effect of time on these global metrics, stratified by remission status (post-treatment MADRS < 11), was assessed using a mixed-effects linear regression model. We found a lower level of local efficiency in depressed patients than control at baseline ( p = 0.0008) in structural networks. No significant difference of baseline network measures were detected in functional networks. Several significant changes following ECT were observed: a decrease in global efficiency (-14% between V1 and V2, p = 0.01) and local efficiency (-15% between V1 and V2, p = 0.01) of functional networks where edges were characterized by the Pearson correlation connecting two regions (Figure 1)
Several significant clinical correlations were observed. The characteristic path length of networks (edges weighted by neurite density index) on the initial MRI (V1) was able to predict clinical remission (MADRS<11) at V3 (p = 0.046). Similarly, the global efficiency (edges weighted by FA) on the initial MRI (V1) was able to predict clinical remission (MADRS < 11) at V3 (p = 0.036); additionally, global efficiency was lower in future remitters at baseline than controls (p = 0.021) (Figure 2). Global metrics of brain network graphs obtained from functional connectivity changed throughout treatment, while metrics from structural connectivity remained stable but differed at baseline between future remitters and non-remitters, as well as between future remitters and healthy controls. These findings suggest that functional metrics may reflect treatment effects, whereas structural metrics may indicate predisposition to ECT response. The global metrics of structural and functional connectivity graphs obtained from diffusion MRI and resting state functional MRI provide potentially relevant non invasive biomarkers for better understanding the mechanisms and predicting the effectiveness of ECT treatment in severe depression.
François RAMON (Paris), Alice LE BERRE, David ATTALI, Clement DEBACKER, Sylvain CHARRON, Maliesse LUI, Marion PLAZE, Catherine OPPENHEIM, Arnaud CACHIA
16:04 - 16:06
#47699 - PG114 Obesity-related reorganization of resting-state brain networks and cognitive correlates.
PG114 Obesity-related reorganization of resting-state brain networks and cognitive correlates.
Obesity has been associated with both cognitive impairment and altered brain function, with widespread alterations in resting-state functional connectivity, particularly affecting networks related to salience, default mode, and executive control (1-4), yet the mechanisms linking these phenomena remain incompletely understood. This study investigates how obesity affects resting-state functional connectivity and brain network topology, and explores how these neural alterations relate to cognitive performance.
We analysed resting-state fMRI data from 115 participants (60 with obesity; 55 controls) using the CONN toolbox and standard preprocessing pipelines. We compared between-group functional connectivity (FC) metrics, including (a) Network-Based Statistics (NBS) -derived network strength, graph theory indices (e.g., global efficiency, degree, path length) (binarized matrices r > 0.2), and (b) Interhemispheric connectivity (IHC) values examined using voxel-mirrored homotopic connectivity (VMHC). Participants also underwent neuropsychological assessment, including California Verbal Learning (CVLT), Wisconsin Card Sorting (WCST), Trail Making (TMT), Digit Span, Stroop, Patient Health Questionnaire-9 (PHQ-9) tests, among others. Partial Least Squares (PLS) identified latent cognitive dimensions significantly associated with group status, and Spearman correlations linked connectivity measures with these cognitive components. Mediation analysis tested whether connectivity metrics mediated the relationship between body mass index (BMI) and the specific cognitive outcomes. Significance thresholds were set to False Discovery Rate (FDR)-corrected p < 0.05. All analyses were adjusted for age, sex, and years of education. The NBS revealed a widespread functional subnetwork showing connectivity changes in those with obesity, involving sensory (e.g., visual, gustatory), reward-related (e.g., accumbens, insula), memory-related (e.g., hippocampus), and executive (e.g., SMA, basal ganglia) regions. Graph theory analyses showed this subnetwork is globally more integrated and efficient in obesity, with shorter average path lengths, higher density, and redistributed centrality. Regionally, the left parahippocampal cortex showed higher degree and efficiency; the precuneus had increased closeness centrality; temporo-occipital regions had reduced eccentricity. In addition, the VMHC analysis identified an increased IHC in the thalamic-caudate region in those with obesity.
The PLS analysis identified three cognitive components significantly associated with obesity: executive function and processing speed (PLS1), verbal memory (PLS2), and a contrast of conceptual reasoning versus memory (PLS3). Positive NBS subnetwork strength was positively associated with both PLS2 (r = .36, p < .001) and PLS3 (r=.32, p<.05). Negative strength (r=-.30, p<.05) and network eccentricity (r=-.29, p<.05) correlated negatively with PLS2. Given the relationship between IHC in the thalamic-caudate region and PLS1, we further explored the association between executive function tests and this measure, which correlated with processing speed (TMTA, r = .27, p < .01). Mediation analysis showed that this IHC in the thalamus/caudate significantly mediated the relationship between BMI and TMT-A (ACME, p=.002), accounting for 52% of the total effect. After including this mediation, the direct effect of BMI on PLS1 was no longer significant Obesity is associated with a reorganization of intrinsic brain network architecture, with global topological shifts and region-specific alterations. Graph-theoretical analyses reveal a globally more integrated, dense, and efficient functional topology in obesity (2, 5). Increased integration of memory- and sensory-related hubs may reflect compensatory or maladaptive mechanisms . Enhanced IHC in subcortical regions may indicate reduced hemispheric specialization or altered sensorimotor integration (6, 7). Connectivity associations with memory and executive function suggest these brain changes underlie cognitive profiles in obesity. The mediation effect of interhemispheric thalamic-caudate connectivity on the BMI–processing speed relationship supports the role of subcortical synchrony in cognitive modulation (7, 8). This study demonstrates that obesity is associated with extensive reorganization of functional brain networks, marked by increased global integration, altered nodal properties in key cognitive regions, and enhanced interhemispheric connectivity within subcortical circuits. These neural alterations are meaningfully linked to specific cognitive domains, particularly memory and executive function, suggesting that the brain’s intrinsic architecture may adapt or maladapt in response to excess body weight. Crucially, the mediating role of thalamic-caudate interhemispheric connectivity in the relationship between BMI and processing speed highlights a potential neural pathway through which obesity impacts cognition.
Elena DE LA CALLE (Girona, Spain), Carles BIARNÉS, Oren CONTRERAS-RODRÍGUEZ, Marian MARTÍ-NAVAS, Victor PINEDA, José Manuel FERNÁNDEZ-REAL
16:06 - 16:51
LIGHTNING TALK POSTER DISCUSSION.
|
Salle Major |
|
"Thursday 09 October"
Added to your list of favorites
Deleted from your list of favorites
D12
15:30 - 17:00
FT1-1 - Basic MR Hardware
FT1: Cycle of Technology
15:30 - 17:00
Basic RF coil design.
Irena ZIVKOVIC (PhD) (Keynote Speaker, Eindhoven, The Netherlands)
15:30 - 17:00
Gradient coils design.
Sebastian LITTIN (Keynote Speaker, Germany)
15:30 - 17:00
Magnet design.
Mark LADD (Keynote Speaker, Heidelberg, Germany)
15:30 - 17:00
Shimming.
Christoph JUCHEM (Keynote Speaker, Vienna, Austria)
|
Salle 120 |
|
"Thursday 09 October"
Added to your list of favorites
Deleted from your list of favorites
E13
15:30 - 17:00
MS1 - How can MRI overcome challenges in multiple sclerosis
|
Salle 76 |
|
"Thursday 09 October"
Added to your list of favorites
Deleted from your list of favorites
G12
15:30 - 17:00
Poster 2
FT3 Poster AI applications | FT1 - Poster AI for image analysis and segmentation | FT3 Poster Post-processing | FT1 - Poster Reconstruction technologies
15:30 - 17:00
#46761 - PG270 Investigate brain volumes with machine learning algorithms to differentiate PD and PSP patients.
PG270 Investigate brain volumes with machine learning algorithms to differentiate PD and PSP patients.
Progressive Supranuclear Palsy (PSP) is diagnosed in the presence of either supranuclear gaze palsy or a combination of slow vertical saccades and postural instability with falls within the first year1. On the same way, postural instability is one of the cardinal signs in Parkinson’s Disease, it is associated with increased falls and loss of independence2. For this reason, PSP and PD, especially in their early stages, show overlapping clinical manifestations3. The aim of the study was to identify the mainly brain regions involved in both diseases to show the differences between PD and PSP diseases using machine learning approach.
In this study, we enrolled 115 PSP from the Neuroscience research center at Magna Graecia University of Catanzaro and 100 PD from the Parkinson’s Progression Markers Initiative (PPMI). All patients underwent a 3T brain MRI scan. We utilized FreeSurfer version 7.1.1 software for automated neuroanatomical segmentation on T1-weighted MRI scans. The "recon-all" function was used to smooth and normalize the images to the "fsaverage" template, reshaping them into an inflated surface. Then, the Desikan-Killiany atlas was applied to divide the cortex into 34 regions of interest (ROIs) per hemisphere. Several morphometric metrics were extracted for each ROI, including cortical thickness (CT), cortical and subcorticall volumes (SV), mean curvature (MC), and estimated total intracranial volume (eTIV). We split the dataset into training and test sets with an 80%–20% ratio. A machine learning (ML) algorithm based on decision trees, specifically XGBoost4, was applied to the entire dataset. Our aim was to differentiate individuals with Parkinson’s disease (PD) from Progressive Supranuclear Palsy (PSP) using morphometric data. To evaluate the model’s performance, we employed nested k-fold cross-validation (n-CV), which consisted of two loops: an outer loop with 4 folds and an inner loop with 4 folds. In the inner loop, we fine-tuned the algorithm and selected the best parameters. The outer loop then estimated the overall performance of the selected model. The final performance metrics were averaged across the different algorithms. The goal of nested CV is to include an algorithm selection step within the inner loop, so any final model to be trained or deployed must go through the same process used in the inner loop (see Figure 1).
Our feature selection approach involved two main steps. First, we evaluated the collinearity of features using correlation analysis, removing any features with a Pearson correlation coefficient greater than 0.6. Next, we trained XGBoost models on the remaining features in each inner fold after hyperparameter tuning. To maximize accuracy, we performed a randomized search (with ten iterations) to optimize parameters like maximum depth, minimum child weight, gamma, number of estimators, subsample, and learning rate. Continuing with feature selection, we assessed feature importance using SHapley Additive exPlanations5 (SHAP). SHAP is a method that explains each prediction by quantifying the contribution of each feature to the model’s output, approximating Shapley values from game theory. Specifically, the SHAP value represents a feature’s contribution to the difference between the actual prediction and the average prediction. In our pipeline, features were ranked based on SHAP importance, and we iteratively removed them to improve accuracy metrics. The performance of the tuned XGBoost model was evaluated based on the mean area under the curve (AUC), accuracy, sensitivity, and specificity in the validation folds of the outer loop. Finally, the best model was tested on the independent test set created initially and on the entire training dataset. Before applying feature selection, the XGBoost classifier using the nested cross-validation method achieved an accuracy of 0.73 and an AUC of 0.80 between PD and PSP patients. After implementing feature selection using SHAP, resulted in an accuracy of 0.82 (AUC 0.87) with 11 selected features (Figure 2). Figure 3 provides useful insights into how specific brain structural measurements impact model predictions. Features such as the left accumbens, left cerebellum white matter, some frontal and temporal thickness, 2 temporal mean curvature and 2 frontal areas had significant effect. This pilot study showed the important difference between PSP and PD, the importance of the results is attributed to the robustness of the nested cross validation method. Another significant aspect of the study is the crucial role of correlation, considering the interrelationship noted among morphometric characteristics. The capability to integrate parameter tuning into the optimization of machine learning models enhances model accuracy while minimizing the risk of overfitting6. Our study aids in the progression of neurodegenerative disease research and emphasizes the significance of using innovative computational approaches to enhance patient care outcomes.
Maria Giovanna BIANCO, Andrea QUATTRONE, Camilla CALOMINO (Catanzaro, Italy)
15:30 - 17:00
#47844 - PG271 MAMU-Net: Boosting nnUNet’s Transfer Learning Performance on Experimental Multimodal MRI Data via a novel Multi-Activation Module.
PG271 MAMU-Net: Boosting nnUNet’s Transfer Learning Performance on Experimental Multimodal MRI Data via a novel Multi-Activation Module.
Accurate segmentation of brain tumors from Magnetic Resonance Imaging (MRI) scans is crucial for diagnosis, monitoring disease progression, and formulating treatment strategies. While traditional methods (RANO & iRANO) are labour-intensive, advanced deep-learning techniques are being explored for more efficient and precise segmentation. In this study, we compare the performance of six state-of-the-art segmentation models: U-Net, nnU-Net, Attention-UNet, ELU-Net, U-Net++ and UNETR [1-7], using the BraTS [8-9] and a private experimental dataset consisting of T2-FLAIR (Fluid-Attenuated Inversion Recovery) and Post-Contrast T1-weighted MRI scans. We evaluate the model performances based on Dice and Jaccard scores [10-11], with nnU-Net consistently achieving the highest accuracy, particularly after transfer-learning on the experimental dataset. To address performance degradation in cross-domain image scenarios, we introduce MAMU-Net by enhancing the base nnU-Net architecture with a novel Multi-Activation Module that fine-tunes the activation layers.
The BraTS datasets contain MRI scans (from the year 2019-2021) from 19 institutions stored in NIfTI format. We've used post-contrast T1-weighted (T1PC) and T2-FLAIR modalities for training different UNet models [Fig. 1] using the hyper parameters as listed in Fig. 1. Expert neuro-radiologists validated manual annotations, marking three regions: Necrotic and non-enhancing tumor core, Enhancing tumor region, and Peritumoral edema. A private MRI dataset of 64 glioblastoma (GBM) patients with 2,413 slices was acquired at the University of Pennsylvania under a collaboration with TCG CREST (data sharing ID: RIS76150) using a 3T Tim Trio MR scanner. High-resolution post-contrast T1-MPRAGE and T2-FLAIR images were coregistered to lower-resolution Diffusion Tensor Imaging (DTI) - B0 images, with a resolution of 128x128x35, as part of a continuation of a project focused on quantitative MR imaging, resulting in lower-resolution images being used for segmentation. Using a semi-automated algorithm, lesions were segmented into Contrast-enhancing (ET), Non-enhancing (NCR), and Edema [12]. This is used as a ground truth. Manual skull stripping was necessary as automated tools struggled with the lower-resolution images. For model optimization, we use Binary Cross-Entropy as the loss function. We evaluate the model's performance using the Dice score and Jaccard Index [13-16].
We aimed to study the impact of different variants of linear unit-based activation functions, i.e. “CELU”, “ELU”, “GELU”, “LeakyReLU”, “PReLU”, “ReLU”, “ReLU6”, “RReLU”, “SELU”, “SiLU”, [17-27] on the best model among “UNet”, “nnUNet”, “Attention UNet”, “ELUNet”, and “UNet++”. The purpose is to enhance performance further. Fig. 3 provides brief details about the activation functions used in this study. We trained a single model in which the activation function is represented as a weighted sum of the ten activation functions described above. We extracted the weights assigned to each activation function and computed their normalized values for each layer. We ensured that each normalized value falls within the range of 0 to 100, and that the sum of all normalized values in each layer equals 100.
We derived our proposed model “Multi-Activation Module Unet (MAMU-Net)”, by modifying the activation function layer of “nnU-Net” and introducing a new Multi-Activation Module in its architecture. The “Multi-Activation Module” is linear combination of “PreLU” and “ReLU6”. Here the coefficients of the linear combination are also trainable parameters. On both datasets, i.e. BraTS validation dataset and the experimental datasets, nnUNet performed best with a Dice score of 88.95% and 64.07% respectively [Fig. 2]. However, the performance of our proposed MAMU-Net was closely aligned with that of nnU-Net. But after transfer learning of last stage, MAMU-Net achieved 74.64% Dice score almost 5% higher than nnUNet (69.86%) [Fig. 2]. After retraining all the decoder stages, MAMU-Net remained best with 85.65% Dice score. Most available open-source datasets consist of pre-operative MRI scans taken before surgery. In contrast, our study evaluates treatment response using post-operative MRI scans, which often include surgical cavities. This poses a challenge in segmentation due to the midline shifting of brain. However, in all types of data nnUNet remained the top performer achieving a Dice score of 88.95% and a Jaccard score of 80.43%. However, it lacked the flexibility to be applied across multiple data sources. Our newly introduced MAMU-NET, featuring the novel Multi-Activation Module, has demonstrated faster feature capture on a new dataset compared to nnU-Net. This makes MAMU-Net more adaptable for domain-specific feature learning. Retraining experiments showed that MAMU-Net achieved the most significant performance improvement, reaching a Dice score of 74.64%, which is 5% higher than that of nnU-Net.
Sankar Narayan MISRA (Kolkata, India), Subhanon BERA, Archith RAJAN, Suyash MOHAN, Harish POPTANI, Sourav BHADURI
15:30 - 17:00
#47898 - PG272 Domain adaptation for MRS deep learning quantification methods.
PG272 Domain adaptation for MRS deep learning quantification methods.
Conventional parametric methods for MRS metabolite quantification suffer from major drawbacks such as modeling biases due to the variability in how prior knowledge, constraints, and parameters are accounted in the fit.
Deep learning methods [1,2,3,4] have also been explored. As most of them rely on a training on simulated data, they might also suffer from modeling biases but this also raise the question of a domain shift when used on real data. A question that remains unaddressed in the context of MRS.
Domain adaptation refers to the challenge of using a model on a test distribution different from the training distribution. Domain adaptation (DA) methods can be either feature based [5,6], data based [7,8] or model based [9,10].
In this work, we propose an experimental protocol to highlight the domain adaptation problem in the context of deep learning for MRS quantification, show which MRS parameters can be a source of domain shift, and then test the efficiency of different DA methods in improving transfer across MRS domains.
In this work, we propose an experimental protocol to highlight the domain adaptation problem in the context of deep learning for MRS quantification, show which MRS parameters can be a source of domain shift, and then test the efficiency of different domain adaptation methods in improving transfer across MRS domains.
We use a simulator based on the Eq. 1, to generate datasets of short echo time (TE=30ms PRESS) MR spectra by randomly sampling values for the amplitude, damping, frequency for each metabolite (simulated with the GAMMA library) and a phase.
Each dataset corresponds to a different domain. We generated 8 different databases (A, B, C… H) of 200000 samples each by combining two different spectral line shapes (lorentzian or gaussian) and four different configurations for macro-molecule profiles. We model the macro-molecule signals as a sum of eleven different macromolecular components, with a base amplitude each, and a variation percentage. The characteristics of each dataset are detailed in Table 1.
For each DA method, we trained one model per domain and tested it on every other domain. A domain shift matrix was then computed to measure the quantification error of each model when it is used each domain. Our goal is to find DA methods to make models perform as well on others domains as on the training domain: we want the domain shift matrix to be as close to zero as possible.
Our quantification model is a convolutional neural network, trained in a supervised fashion. It is composed of two parts: an encoder composed of four convolutional layers, each followed by a batch normalization layer, an activation function and a sub-sampling, and a regression head composed of two fully connected layers. The output of the model for a spectra is a vector composed of the amplitude, damping factor and frequency shift of each metabolite and a global phase. Our baseline is this model without any DA method.
We then combine this model with the test-time batch normalization (TTBN) [11]. This method replaces the mean and standard deviation computed on the training dataset of each BatchNorm layer by those recomputed on the test dataset. Values in the domain shift matrix are the logarithm of the MSE of the amplitude between the predicted values and the ground truth. Each line correspond to a training domain and each column to a test domain. The matrix was normalized by subtracting the diagonal (same train and test domain) to each line.
In fig. 2, the matrices "Base model" and "TTBN" respectively show the quantification errors for the base model and the TTBN. For the base models, best results are mainly obtained on the diagonals. This is the expected result, as a model should indeed perform better when applied on data similar to those that it was trained on. Performance is degraded in other cases. Models A, B, E and F, that were trained with lower macro-molecule amplitude variations, perform worse when tested on domains with different macro-molecule base amplitudes, while models C, D, G and H appear to suffer less from the domain shifts.
Regarding the TTBN method, even though the domain shift metric is still mostly the lowest on the diagonals, the matrix is more homogeneous, and the performance on other domains are less degraded. This shows that the TTBN method improves indeed transfer across domains. First, one can see that the performances of deep neural networks for MRS quantification can be altered by a domain shift: the macromolecular profiles and the spectral line shapes can be a source of domain shift between databases, but other parameters have to be considered, and might also be a source of data distribution shift.
Second, we show the importance of considering DA methods. TTBN appeared to be efficient in improving transfer across domains. Other DA methods from the literature should also be tested, and the design of DA methods that would suit the specificities of MRS signals should also be considered.
Slim HACHICHA (Lyon), Michaël SDIKA, Hélène RATINEY
15:30 - 17:00
#47880 - PG273 Deep learning-based generation of a digital MRI brain phantom.
PG273 Deep learning-based generation of a digital MRI brain phantom.
The widespread use of neural networks for MRI data analysis and the limited availability of real data for training underscore the need for synthetic MRI datasets [1, 2]. To generate these synthetic datasets, digital phantoms are essential. A digital MRI phantom (in the basic version) comprises tissue characteristics such as T1 and T2 relaxation times and proton density (PD) [3]. These phantoms simulate the properties of various tissues during MRI acquisition [4]. We aim to investigate the capabilities of neural networks, particularly diffusion models, for generating digital MRI phantoms from weighted MR images. The novelty of our study lies in the simultaneous generation of three quantitative maps: T1, T2, and PD.
A) Dataset.
The training dataset consists of quantitative maps of T1, T2 relaxation times, and proton density (PD), along with corresponding synthetic MR images weighted for T1, T2, and PD. Thus, the dataset contains 3482 pairs of quantitative maps and weighted images. These quantitative maps were generated using the methodology outlined in [5], employing anatomical brain models from the BrainWeb database [3]. These models include 11 distinct tissue types, each assigned specific T1, T2, and PD values sourced from the literature [4, 6, 7]. To introduce variability and prevent uniformity, tissue parameter values within each anatomical region were sampled from a normal distribution centered around the mean values, with a standard deviation of 10%. To obtain synthetic weighted images, we developed a pipeline, illustrated in Figure 1. The process involves three key steps: generating digital phantoms from quantitative maps (T1, T2, PD), simulating MR signals using Bloch’s equations (via the KomaMRI software package [8]), and reconstructing the final MR images. In this study, employed pulse sequences were T1-, T2- and PD-weighted 2D turbo spin echo (TSE) (TR = 700ms/3.0s/6.0s, effective TE = 7.65ms/76.5ms/6.69s, ETL =10, resolution – 1.95*1.95*5mm^3, field-of-view – 0.25*0.25m), constructed using the open-source Pulseq framework [9]. KomaMRI also requires T2* maps for input, in our case they were replaced with T2 maps, because sequences based on TSE are generally not sensitive to T2* effects.
B) Model.
Denoising Diffusion Probabilistic Models (DDPMs) excel in generating high-fidelity images and are widely used in computer vision [10]. However, their reliance on extensive diffusion steps results in prolonged training and sampling times [11]. To address this, we adopted the Fast Denoising Diffusion Probabilistic Model (Fast-DDPM), which achieves comparable image quality with fewer than 1000 time steps [11]. During training, the model uses quantitative T1, T2, and PD maps as inputs, with T1-, T2-, and PD-weighted MR images serving as conditioning data. At inference the model takes weighted MR images (as the condition) and Gaussian noise (as input) to predict the underlying quantitative maps. Our implementation used 800 diffusion steps with a uniform noise scheduler. Training was conducted over 763 epochs with a batch size of 8, and the image size was 128 × 128 pixels. Figure 2 illustrates the training and inference process of the model.
C) Evaluation.
The test set consisted of 100 brain slices from a separate anatomical model (T1, T2, PD-weighted images). To evaluate the model's quality, metrics such as the Structure Similarity Index (SSIM) [12] and Peak Signal-to-Noise Ratio (PSNR) [13] were used. The trained Fast-DDPM successfully generated quantitative T1, T2, and PD maps. To assess their quality, Figure 3 provides a direct comparison between the ground truth and synthetic maps. Figure 4 shows the probability density curves for the original and synthesized maps. For the T1 quantitative maps SSIM achieved 0.78±0.05, and PSNR - 22.12±0.55 (average of 100 slices). For T2 quantitative maps SSIM – 0.79±0.02, PSNR – 18.67±0.45, for PD quantitative maps SSIM – 0.55±0.04, PSNR – 19.32±0.35. The synthetic phantoms produced by our method exhibit high structural similarity to the original ground truth for T1 and T2 maps, and moderate for PD maps. Notably, our framework simultaneously generates all three quantitative maps (T1, T2, and PD) without relying on prior segmentation. To date, we have been testing the model on synthetic data from a separate anatomical model. In the future, we plan to test the model on real weighted MR images and real ground truth quantitative maps. The DDPM-based approach shows potential for creating realistic digital phantoms. The creation of realistic brain phantoms holds significant promise for advancing MRI research - for pulse sequence and reconstruction algorithms design, and for generation of synthetic MRI datasets.
Acknowledgements
This work was supported by the Ministry of Science and Higher Education of the Russian Federation (Project FSER- 2025- 0009).
Kseniya BELOUSOVA (St. Petersburg, Russia), Zilya BADRIEVA, Iuliia PISAREVA, Nikita BABICH, Dmitriy AGAPOV, Olga PAVLOVA, Ekaterina BRUI, Walid AL-HAIDRI
15:30 - 17:00
#47631 - PG274 Assessing generalizability in choroid plexus automatic segmentation: a comparison of ASCHOPLEX fine-tuning and Dafne federated incremental learning.
PG274 Assessing generalizability in choroid plexus automatic segmentation: a comparison of ASCHOPLEX fine-tuning and Dafne federated incremental learning.
The Choroid Plexus (ChP) is a vascular brain structure involved in vital brain functions [1,2]. Most clinical studies focused on analyzing ChP Volume (ChPV) in diseased populations, hypothesizing a link between increased ChPV and neuroinflammatory processes, although this association is influenced by multiple factors [3,4]. ASCHOPLEX [5], a deep learning-based toolbox with an integrated fine-tuning step, returns accurate ChP segmentations on non-contrast-enhanced T1-weighted MRI images [6], but struggles with data variability [7]. To enhance its robustness, this work integrates ASCHOPLEX into Dafne [8], a federated incremental learning framework that supports privacy-preserving model updates [9,10]. In contrast to classical fine tuning, Dafne enables adaptation to new data while retaining previously acquired knowledge by performing a weighted averaging of the pre-existing and the fine-tuned model at every aggregation. This study aims to evaluate the generalizability of ASCHOPLEX by comparing the performance of fine-tuning and federated learning strategies across two Multiple Sclerosis (MS) cohorts.
Details of the data are provided in Tab.1. Dataset 1 (D1) consists of 128 subjects acquired at the Multiple Sclerosis Centre of the University Hospital of Verona, Italy [5]. Dataset 2 (D2) includes 19 MS subjects from the publicly available ISBI 2015 Longitudinal MS Lesion Segmentation Challenge, including the first time point for each subject [11,12]. ASCHOPLEX architectures were modified by incorporating the following models implemented in MONAI [13]: DynUnet [14], UNETR [15], and SwinUNETR [16]. A five-fold cross-validation training was performed on D1. Training parameters and data augmentation techniques were consistent with those described in [5], with the addition of data augmentation transforms from TorchIO [17]. The five best-performing fold configurations were selected based on the Dice Coefficient and combined using an ensemble majority voting strategy. These models were then separately fine-tuned following the same procedure as in [5], and integrated into Dafne [8] to enable incremental learning on D2 using 400 epochs. Dafne was adapted to support 3D models in MONAI, including ensemble processing and server-side model merging. Three prediction strategies were evaluated on the test sets of both datasets: direct prediction following initial training on D1 (DP), prediction after fine-tuning on D2 (FT), and prediction following incremental learning on D2 (IL). The predicted segmentations were compared to expert-generated ground truth (GT) manual segmentations. Segmentation performance was assessed using absolute ChPV, Dice Coefficient, 95th percentile Hausdorff Distance (95% HD), and Percentage Volume Difference (ΔVol%), using Python (v 3.12). Selected configurations include two UNETR models, a DynUNet model, and two SwinUNETR models with a patch size of 128 and loss function combining Generalized Dice and Cross-Entropy. The segmentation metrics (Tab.2, Fig.1) on DP report a drop in performance on D2 compared to D1 (mean Dice/ΔVol%: D1=0.80/8.96%, D2=0.35/-71.53%). In contrast, FT and IL approaches on D2 show improvements compared to DP (mean Dice: FT=0.56, IL=0.43), despite ΔVol% consistently exceeding 55%. Concerning the retesting on D1 after ASCHOPLEX’s exposure to D2, FT performance drops significantly compared to DP (mean Dice/ΔVol%: 0.12/-89.02%), with 95%HD considered unreliable. Meanwhile, the IL approach yields ChPV estimates that are closer to DP (Δ(ΔVol%): IL-DP= -1.8%, FT-DP=-98%). Fig.2 displays the compared segmentations overlaid on the T1-weighted MRI for a representative subject from both D1 and D2. This study investigates the generalizability of fine-tuning and federated incremental learning approaches for the ChP segmentation task. The findings indicate that fine-tuning deep learning models on a new dataset enhances performance compared to direct prediction using a model trained on a different dataset. However, this improvement comes at the cost of an irreversible loss of information from the dataset used for initial training, mostly when datasets have different image intensity range (Fig.2), as demonstrated by the performance degradation observed when re-evaluating the fine-tuned model on the D1 test set. In contrast, the federated incremental learning strategy implemented within the Dafne framework yields comparatively lower performance but offers greater generalizability. Specifically, merging the model trained on D1 with a model briefly trained on D2 improves prediction accuracy on D2 relative to direct prediction, while preserving the knowledge previously acquired from D1. To conclude, integrating ASCHOPLEX into the federated incremental learning Dafne framework enables better generalization across datasets by preserving prior knowledge, offering a robust alternative to fine-tuning for ChP segmentation in MS studies.
Valentina VISANI (Padova - Basel, Italy), Mattia VERONESE, Agnese TAMANTI, Francesca Benedetta PIZZINI, Massimiliano CALABRESE, Marco CASTELLARO, Francesco SANTINI
15:30 - 17:00
#47877 - PG275 Neural network approach to T2 mapping based on multi-echo turbo spin-echo: training on a generated dictionary.
PG275 Neural network approach to T2 mapping based on multi-echo turbo spin-echo: training on a generated dictionary.
Multi-echo turbo-spin echo (ME-TSE) is a fast MRI pulse sequence used for T2 mapping, offering significant speed advantages over alternatives like multi-echo spin echo (MESE). This efficiency is critical for breath-hold scans or dynamic studies. Dictionary-based T2 mapping (Echo Modulation Curve - EMC method) has proven effective for ME-TSE and MESE data, but is computationally intensive [1, 2, 3], as it requires direct matching of pixel curves and the entire dictionary. The original EMC method authors reported a reduction in fitting time by implementing a gradient descent (GD) optimization scheme [4].
An alternative acceleration strategy combines neural networks (NN) with the EMC method. For instance, a U-Net trained on brain MESE data successfully replicated EMC-derived T2 maps [5]. Shifting from image-based to dictionary-based training could improve robustness across different scan areas, as shown for upper leg T2 mapping [6]. Yet, that approach simulated echo modulation curves using extended phase graph (EPG) formalism, which does not account for slice-profile effects. Moreover, EPG’s computational complexity escalates exponentially with longer pulse sequences.
While neural networks have been applied to MESE, their adaptation to ME-TSE, where echo modulation curves typically comprise only ~3 values (vs. many in MESE), remains unexplored. Here, we investigate a neural network-based T2 mapping method for ME-TSE, trained on a dictionary of Bloch-simulated echo modulation curves.
Agarose-based phantoms (Eurospin) with calibrated reference values (T1ref= 336.1/672.5/1235.2/1288.9/1407.7 ms and T2ref= 97.2/118.5/170.5/158.7/131.6 ms) were scanned at 3T (Magnetom Vida, Siemens). The ME-TSE pulse sequence parameters were: TR = 1.5 s, first TE = 7.4 ms, inter-echo time = 7.4 ms, turbo factor = 3, number of contrasts = 3. Additionally, an in vivo scan of the lower leg muscle was acquired using identical parameters, except for TR = 2 s. In ME-TSE, k-space for each image is filled with data acquired at different echo times, where the number of such echoes is defined as the turbo factor. We employed an effective TE combination identified in our previous work [7].
Using a Matlab script [3], five EMC dictionaries were generated for each phantom with parameters: B1+ = [80:1:120] %; T2 = [50:1:240] ms; T1 = T1ref of phantom. For muscle one EMC dictionary were generated with next parameters: T2 = [5:0.25:100] ms; T1 = 1150 ms. The dictionaries were augmented with Rician noise based on SNR characteristics estimated from MR images. Training data size after augmentation for each phantom: 15662 curves, for muscle: 31242 curves.
Similar to [6], a fully connected neural network (multilayer perceptron) was selected using the Keras library. The training and inference pipeline for the neural network are presented in Figure 1. Initially, T2 maps were reconstructed using the EMC dictionary method. After that, T2 maps were generated based on the neural network illustrated in Figure 1. In Figure 2A, T2 maps obtained using various methods are presented. Visually, the maps produced by the EMC method and the NN method appear similar; however, the T2 maps for the NN method exhibit less noise. Figure 2B shows a correlation plot between the T2 values of each pixel inside region of interest for both methods, with calculated Pearson coefficient. Figure 2C displays the mean relative errors compared to the reference T2 values for both methods. It can be seen that both methods have approximately the same error, however, NN-method demonstrates lower standard deviation of the error.
The T2 maps for muscle tissue obtained using the two different methods also show no significant difference (Figure 3A), as can be seen in the difference map of the two (Figure 3B). Figure 3C presents a correlation plot for T2 values of each pixel in five regions of interest for the muscle (Figure 3D). A high correlation with a strong Pearson coefficient is observed. In this study, we demonstrate the feasibility of a T2 mapping method based on neural network trained on simulated echo modulation curves obtained using Bloch simulations for a multi-spin turbo spin-echo pulse sequence. A high correlation between the results of using NN and EMC methods were observed. Additionally, the NN fitting took nearly 13 times less time than the classic EMC method, requiring only 1s compared to 12.87s for a 66x197 matrix. In the future, the NN method could be used for multi-slice T2 mapping and could also be adapted to estimate another parameters, such as T1. In conclusion, it has been demonstrated that the use of neural networks can effectively replace the EMC dictionary-matching method, resulting in lower T2 estimation errors and higher processing speeds. By employing Rician noise augmentation and training the data with low T1-weighting, we achieved a very low median relative error in T2 estimation.
This study was supported by the Russian Science Foundation (RSF) grant No. 23-75-10045.
Zilya BADRIEVA (Saint Petersburg, Russia), Anna KONANYKHINA, Ekaterina BRUI
15:30 - 17:00
#47831 - PG276 Self-supervised learning of human spinal cord anatomy - SpineGPT.
PG276 Self-supervised learning of human spinal cord anatomy - SpineGPT.
Personalized spinal cord models are essential for enhancing the effectiveness of epidural electrical stimulation (EES) therapies aimed at restoring neurological functions in individuals with spinal cord injury (SCI)[1–6], and other diseases [7, 8]. Built on detailed, patient-specific medical imaging data, these models play a critical role in both the pre-operative planning and post-operative fine-tuning of EES implant placements.
The processing and annotation of medical imaging required to create the spinal cord personalized model is labor-intensive and time-consuming[3]. Anatomical tissue segmentation is a bottleneck for the scalability and widespread clinical adoption of personalized spinal cord models.
Artificial neural networks (ANNs) hold promise for automating tissue segmentation from medical imaging, thereby reducing reliance on manual intervention. ANNs have proved successful in many segmentation tasks[9]. However, they face challenges due to the scarcity of annotated medical data and the variability between patients, imaging protocols, anatomical regions, and scanner types[10, 11].
Self-supervised learning (SSL) has emerged as a powerful method for deriving meaningful representations without human supervision, leveraging available unlabeled data. SSL has fueled the success of natural language processing (NLP) models like ChatGPT[12], which learn the structure and semantics underlying language by predicting masked (hidden) elements in sequences of words based on the surrounding context. This self-supervised objective enables NLP models to perform downstream tasks such as text summarization with remarkable efficiency.
Taking inspiration from NLP, we applied SSL methodologies to unlabeled spinal MRI volumes to develop generalistic models that can be adapted to downstream tasks with minimal expert intervention. Our framework, SpineGPT, learns by reconstructing masked parts of the MRI volumes from their visible counterparts, extracting representations that capture the spinal cord’s underlying structure. We use these representations to fine-tune SpineGPT to solve clinically-relevant problems such as tissue segmentation.
Our current implementation of SpineGPT builds on a masked auto-encoder (MAE) framework[13], learning self-supervised representations of MRI scans through a self-reconstruction objective. In this process, a 3D chunk of the scan is largely masked (80%) at random and used as input to an autoencoder neural network, which is trained to reconstruct the missing portions from the visible parts (Figure 1). Through this pretraining phase, the model’s encoder learns to map the original volume to a latent representation that captures informative structural details.
SpineGPT’s current encoder and decoder consist of standard Vision Transformer (ViT) layers adapted for 3D volumes. We leverage all available unlabeled data, totaling over 1,300 diverse T2 weighted MRI volumes of the spine from both publicly available[10, 14–17] and private datasets[3, 4, 6–8, 18].
Segmenting spinal tissues such as spinal roots, white matter (WM), and cerebrospinal fluid (CSF), is vital for developing personalized spinal cord models. Current methods, based on supervised learning, allow for segmentations for tissues except spinal roots in the lumbosacral region[19–23].
We propose a strategy of freezing the encoder of SpineGPT and finetune the decoder to segment previously described spinal tissues. The input volumes are cropped around the spinal cord[21, 24] into chunks of 32x32x64 with patch sizes of 4x4x8. The current architecture of the encoder of SpineGPT consists of 12 ViT layers with 16 self-attention heads[25, 26]. The decoder consists of 8 ViT layers. The pre-trained phase minimizes the reconstruction error between the input patches and the original patches using Mean Square Error (MSE).
We finetuned the decoder on the available labeled datasets containing CSF, WM and spinal roots [3, 6–8] consisting of 31 individual T2 weighted high resolution MRI (Resolution 0.3x0.3x0.6 mm3). We minimized the Dice loss[27]. We tested our segmentation framework on two test sets. First, we tested on a subset (2 individuals) containing both dorsal and ventral spinal roots. Secondly, we tested on a public labeled data set of 14 participants on the lumbosacral region[28] only containing dorsal roots. We achieved promising preliminary results, with Dice scores of 0.95±0.01, 0.94±0.01 and 0.73±0.00 on the first test set (Figure 2) and 0.91±0.03, 0.89±0.23 and 0.41±0.17 on the second test set for segmentations of CSF, WM and spinal roots respectively. We introduce SpineGPT, a foundation framework for spinal cord T2 weighted MRI. We finetuned SpineGPT to address, for the moment, one downstream task, lumbosacral spinal tissues segmentation, in particular spinal roots. Preliminary results are promising, achieving state-of-the-art results in the segmentation task. However, extensive testing is still necessary to ensure a robust framework.
Sergio Daniel HERNANDEZ CHARPAK (Lausanne, Switzerland), Icare SAKR, Léon MULLER, Juliette HARS, Jonas BLANC, Bilel EL-GHALLALI, Philippe FORERO, Jean-Baptiste LEDOUX, Fabio BECCE, Jocelyne BLOCH, Grégoire COURTINE, Henri LORACH
15:30 - 17:00
#46441 - PG277 T1-PILOT: Optimized Trajectories for T1 Mapping Acceleration.
PG277 T1-PILOT: Optimized Trajectories for T1 Mapping Acceleration.
Cardiac MRI T1 mapping quantifies the T1 relaxation time of cardiac tissues, providing critical insights into tissue composition and aiding in diagnosing pathologies such as fibrosis, and diffuse myocardial inflammation. Data acquisition requires multiple breath-holds and is sensitive to motion artifacts, limiting feasibility for high-resolution imaging. Compressed Sensing has shown success in accelerating data acquisition, however most existing methods do not consider the physical exponential decay model in optimization, and rely on fixed undersampling patterns that do not fully exploit acceleration potential.
Our pipeline is composed of 3 conceptual layers (see attached figure) - first, a subsamlping layer, that models a learned, per-frame parametric non-Cartesian acquisition set,and applies it over the input data. Second, a neural reconstruction block, performing denoising before decay estimation, to better-inform both the decay estimation model in the forward pass and the samplng scheme in the backward pass. Lastly, we use a decay estimation model, which a neural network that outputs exponential decay parameters. These parameters are used to perform exponential regression, and the residuals of this regression are used as a loss objective that is backpropagated through all 3 layers. For acquisition parameterization we follow [1], for the reconstruction and decay models we use the reconstruction model from [2].
To efficiently leverage data-priors for learning an acquisition set that would generalize well across multiple sequences, we first optimize a non-Cartesian acquisition set solely for reconstruction (train only the sampling and reconstruction layers). At a second stage, we finetune this set for decay estimation, guided by the physical decay model. Finally, we propose finetuning the reconstruction and decay models per-sample for optimal estimation results. The core of our work is showing the learned, per-frame learned acquisition schemes, alongside the inclusion of the physical decay model in optimization, greatly benefit T1 Mapping acceleration. To show this, we compare T1-PILOT to four baselines that are representative of current approaches for acquisition and learning objectives used in training – radial acquisition (constant sampling scheme shared across frames) , GAR acquisition (constant per-frame sampling), a single, learned acquisition pattern shared across frames, and per-frame learned acquisition not trained under the physical decay model. T1-PILOT significantly outperformed baseline strategies, achieving higher T1 map fidelity at greater acceleration factors. Specifically, it demonstrated consistent gains in Peak Signal-to-Noise Ratio (PSNR) and Visual Information Fidelity (VIF) relative to existing methods, along with marked improvements in delineating finer myocardial structures. We demonstrate the advantage of incorporating physically feasible, per-frame non-Cartesian learned acquisition schemes in T1-Mapping acceleration. We also show that intergrating the physical decay model directly within the learning objective dramatically improves acceleration potential. This approach addresses the limitations of traditional methods that rely on fixed undersampling patterns and demonstrates the potential of physics-informed deep learning in cardiac MRI.
We hope these findings would encourage and inform future endeavors in T1-Mapping and general MRI acceleration.
One limitation of our work is that the multiple optimization steps require higher computational resources, in comparison with current methods. We aim to explore alternative optimization approaches that would alleviate this difficulty. Another limitation is that our finidngs mostly rely on simulated data created from fully-sampled k-space. We aim to test our method on actual MR machines in the future. The integration of the T1 signal relaxation model into the sampling-reconstruction framework allows T1-PILOT to optimize k-space trajectories effectively, leading to enhanced quantitative accuracy and reduced acquisition times. This approach addresses the limitations of traditional methods that rely on fixed undersampling patterns and demonstrates the potential of physics-informed deep learning in cardiac MRI.
Tamir SHOR (Haifa, Israel), Moti FREIMAN, Chaim BASKIN, Alex BRONSTEIN
15:30 - 17:00
#47770 - PG278 Enhancing Conventional CNNs with Compound Scaling for Brain Tumor Detection in MRI.
PG278 Enhancing Conventional CNNs with Compound Scaling for Brain Tumor Detection in MRI.
Cancer remains the primary cause of mortality globally [1], underscoring the need for leading-edge diagnostic tools. Magnetic Resonance Imaging (MRI) is a non-invasive, frequently used modality in brain tumor detection as it provides insights of clinical importance, ensuring early brain tumor detection [2]. MRI offers precise identification of tumors by contrasting soft tissue without ionizing radiation [3].
Machine learning based diagnosis systems have demonstrated enhanced diagnosis and classification results by utilizing advanced CNNs (Convolutional Neural Networks) for MR images. For instance, Residual fused shepherd CNN [4] is a hybrid model that provided accuracy of 94% in brain tumor detection. In [5], Binary classification of brain tumors has been explored using several CNN variants, presenting the performance analysis of Alexnet, VGG-16, GoogleNet and RNN. The evaluation on BRATS 2013, BRATS 2015, and OPEN I dataset revealed that AlexNet achieved the highest accuracy of 98.7%.
Based upon the insights from literature, it is evident that CNNs has a very crucial rule for brain tumor detection in MR images. A most important advancement in CNN optimization is EfficientNet [6], which employs compound scaling to balance network depth, width, and resolution, to improve model efficiency and accuracy. Therefore, it is worthwhile to analyze its impact in comparison with conventional architectures.
The main objective of this study is to analyze the impact of EfficientNet’s compound scaling method on conventional VGG-16, VGG-19 [7], ResNet-50, ResNet-101 [8], and DenseNet-121, DenseNet-169 architectures [9].
In this study, the compound scaling strategy of EfficientNet is applied to each of the conventional architecture as explained in the introduction section and the performance of unscaled conventional models is compared with their respective scaled versions. Firstly, a customized dataset is made by combining the datasets created from different sources comprising MR scans, that is Figshare [10] and Kaggle sources [11],[12]. The dataset is categorized into four tumor classes (glioma, meningioma, pituitary tumor and no tumor) and preprocessed through resizing (130×130 for conventional and 156×156 for enhanced compound scaled variants) followed by normalization.
The EfficientNet model width scaling in conventional CNN is represented by Eq.1:
w′ = w * β^∅ (Eq. 1)
The resolution scaling is represented by Eq.2:
r′= r * γ^∅ (Eq. 2)
Where parameters in above equations are;
w: original width (number of filters),
r: original input resolution,
β: width scaling factor,
γ: resolution scaling factor,
∅: compound coefficient,
w′, r′: scaled width and resolution, respectively
The value of compound coefficient ∅ is set to 1.2 as explained in EfficientNetB2 [6]. The compound scaling is applied in width and resolution only as shown in Figure 1. The depth of conventional CNNs remains constant as each architecture is designed with a predefined depth factor. All models are trained with 80-20 training-testing split, using categorical cross-entropy loss function and Adam optimizer. Performance metrics include accuracy, precision, recall, and F1-score. The impact of EfficientNet compound scaling on CNN architectures is evaluated using key performance metrics including accuracy, precision, recall and F1-score. Table 1 shows the model comparisons and highlights the impact of compound scaling on different CNNs architecture performance.
The best accuracy results were achieved by DenseNet-121(90.21% unscaled, 95.81% scaled), followed by scaled VGG-16 and VGG-19 at 94.68%. ResNet-101 exhibited a substantial accuracy gain, rising from 73.24% to 90.84% after scaling. ResNet-50 demonstrated relatively low accuracy (77.13% unscaled, 81.83% scaled). The analysis of proposed scheme across all models shows that the most prominent improvement from compound scaling is observed in ResNet-101, with accuracy increasing by over 17%. Overall, the proposed strategy for scaling the conventional models demonstrates better generalization and fast convergence.
Figure 2 demonstrates the performance of each model in classifying brain MR images for tumor predictions, under unscaled and scaled conditions. All the models have demonstrated strong performance in tumor classification after applying compound scaling.
The future research could focus on refining scaling parameters and exploring more effective data augmentation techniques to further enhance model performance. The results clearly indicate that the EfficientNet compound scaling strategy significantly improved accuracy of conventional models. This highlights the substantial impact of architecture choice and scaling on the performance of conventional CNNs in MRI based brain tumor detection. Although the effectiveness of this method varied across models, the EfficientNet scaling method enhances the feature learning and overall classification performance.
Muhammad Hassan FARID (Islamabad, Pakistan), Muhammad Adnan NASIM, Muhammad Abdullah UMAR, Usman JAVED
15:30 - 17:00
#47684 - PG279 Glioseg: an integrated framework for robust AI-based glioma segmentation.
PG279 Glioseg: an integrated framework for robust AI-based glioma segmentation.
Treatment planning and surveillance of patients with glioma can benefit greatly from accurate tumor segmentation [1,2]. However, manual segmentation is time-consuming and subject to intra- and inter-rater variability [3]. While recent advances in artificial intelligence (AI) have yielded automated segmentation tools with expert-level performance [4], many existing tools rely on specific preprocessing steps or consist of separate installable components, complicating their use in clinical research settings [5]. We present Glioseg, a fully automated, end-to-end framework for multi-compartment glioma segmentation that integrates robust preprocessing, multiple AI models, and ensemble-based label fusion. The framework is designed for easy deployment with minimal configuration.
Glioseg requires four pre-operative structural MRI scans, including T1-weighted (T1w), contrast-enhanced T1-weighted (T1CE), T2-weighted (T2w), and T2-weighted fluid-attenuated inversion recovery (FLAIR) MRI, provided in DICOM or NIfTI format (Figure 1). Preprocessing steps include (in order) reorientation and resampling to a standard space and resolution, intra-subject groupwise registration, registration to an appropriate brain atlas, and brain extraction. Five publicly available pre-trained AI models are used for multi-compartment tumor segmentation, targeting non-enhancing tumor/edema, necrosis and enhancing tumor. The specific models used are nnUNet [6], the winning submission of the BraTS 2021 challenge [7], HD-GLIO [6,8], DeepSCAN [9] and the Federated tumor segmentation (FETS) tool [10,11]. The choice of brain atlas for registration (MNI152 [12] or SRI24 [13]) is determined by the input requirements of each model. Segmentation outputs are postprocessed, including isolated voxel removal and volume-based relabeling, followed by ensembling through simultaneous truth and performance level estimation (STAPLE) [14]. Final segmentations are transformed back to native patient space to facilitate downstream analysis. Glioseg was evaluated on an external dataset of 43 patients with adult-type diffuse glioma of varying WHO CNS5 (2021) [15] types and grades. All scans completed the pipeline without failure. Segmentations were compared in native space to semi-automatic ground truth annotations, where outputs from an external AI model were corrected by radiologists. Glioseg achieved an average whole tumor Dice score of 0.85 ± 0.09 and a multi-class Dice score of 0.77 ± 0.12 (Figure 2). The average whole tumor Dice scores of the individual models ranged from 0.78 to 0.86, while multi-class Dice scores ranged from 0.70 to 0.80. Figure 3 shows a representative case with high segmentation agreement, while Figure 4 highlights a common segmentation error seen, in which extensive non-enhancing tumor/edema is mislabeled as necrosis. Glioseg addresses several limitations of current segmentation tools by supporting multiple scan formats, automating pre- and postprocessing, and running five models end to end in a single workflow. However, the STAPLE ensemble did not outperform the strongest individual model (BraTS ‘21), suggesting that the current ensembling strategy may not fully leverage complementary model strengths and could be hindered by lower-performing models. Future work will explore alternative label fusion methods, such as reliability-weighted ensembling or leave-one-out strategies to identify and exclude detrimental models. A persistent error was the misclassification of non-enhancing tumor/edema as necrosis. This issue is likely an inherent limitation of the individual models, which were primarily trained on whole-tumor, tumor-core, and enhancing-tumor labels without explicit differentiation between necrosis and non-enhancing tumor. Our current postprocessing only partially mitigates this problem. To improve, we will explore intensity-based or structural constraints, such as enforcing a rule that necrosis must be encapsulated by enhancing tumor. Additional ongoing work includes benchmarking against fully manual expert segmentations and established tools in the literature, stratifying performance by tumor type and size, and expanding the pipeline to include post-operative segmentation models and models that are robust to missing structural scans. Glioseg provides a fully automated solution for clinically meaningful multi-compartment glioma segmentation. Its combination of standardized preprocessing, multi-model inference, and ensemble-based fusion in a single framework ensures reliability and user-friendliness. Ongoing development aims to further enhance performance and facilitate reproducible segmentation outcomes. With continued validation, Glioseg has the potential to become a standard tool to support glioma research.
Juancito Cc VAN LEEUWEN (Rotterdam, The Netherlands), Gonzalo E MOSQUERA ROJAS, Ivar Jhg WAMELINK, Vera Cw KEIL, Marion SMITS, Stefan KLEIN
15:30 - 17:00
#45515 - PG280 Automated MS Lesion Detection with NeuroQuant: Comparing the Hybrid Machine and Deep Learning Version 4 With Its Predecessor.
PG280 Automated MS Lesion Detection with NeuroQuant: Comparing the Hybrid Machine and Deep Learning Version 4 With Its Predecessor.
NeuroQuantⓇ MS (NQ-MS, formerly LesionQuant) is a medical software for detecting, counting, and measuring multiple sclerosis (MS) lesion volumes seen as hyperintensities in T2w-FLAIR images, categorizing them by brain regions–(cortico)juxtacortical, periventricular, infratentorial, and deep-white-matter–based on McDonald criteria[1], while also calculating lesion burden. In 2020, Brune et al.[2] from our MS research group in Oslo found NQ-MS useful in aiding visual MS lesion detection and improving brain atrophy identification, the latter being beyond the scope of this study. More recently, our NQ-MS software was upgraded from version 3.1 to 4.12, integrating a hybrid machine learning (ML)/deep learning (DL) algorithm for enhanced segmentations[3].
This study compares the two versions using a locally collected multiple sclerosis (MS) dataset, hypothesizing that ML/DL segmentation improves lesion detection, assessed visually and through statistical analysis.
Sixty-three subjects with MS were scanned on a 1.5T Siemens Aera MRI scanner using a standard 20-channel head coil, with T2w-FLAIR and T1w-MPRAGE (for brain segmentation) and image parameters closely following NQ-MS recommendations. The dataset is part of a larger study approved by the hospital’s local ethical committee.
Lesion detection volume was set to a minimum of 3 mm³ with 1 mm separation[2]. In NQ-MS version 3.1, sensitivity thresholds (range: 0–2, high to low) were applied. First the default of 1.0 (neutral), then adjusted to 1.2 (neutral-) for periventricular lesions to avoid over-detecting non-lesion structures. Visual assessments of some patients have been done, whereas all the segmentations will be reviewed by two radiologists in time for the conference. Statistical analysis of expert versus program analysis will be done using Python 3 with the Pandas module[4]. Cohen’s Kappa results will be analyzed for lesion classifications, and ICC/CCC for lesion filling percentages in NQ-MS segmentation masks. Statistical analyses of the lesion counts and volumes were conducted using R Software (version 4.3.0)[5]. The comparison of performance between the two NQ-MS versions was implemented through Wilcoxon signed-rank tests for each brain region. Preliminary analysis of expert assessment versus NQ-MS segmentation shows a significantly larger agreement between expert and v4.12 than its predecessor. This pertains both to lesion type classification and lesion filling grade. There was also a significant preference by the expert for v4.12 segmentation.
Figure 1 presents boxplots of lesion counts and volumes for the two NQ-MS versions, demonstrating statistically significant increases in v4.12 compared to v3.1, except for periventricular lesion counts. As the NQ-MS v3.1 manual suggests that a sensitivity of 1.2 may be more appropriate for detecting periventricular lesions – since a setting of 1.0 might capture non-lesion tissue structures – we reprocessed the dataset with this adjusted sensitivity. The corresponding results are shown in Figure 2. Our study demonstrates that upgrading from NQ-MS v3.1 to v4.12, which incorporates ML/DL, results in a significantly higher sensitivity in lesion detection and volumetry as evaluated with statistical tests. NQ-MS v4.12 consistently detected more brain lesions and with larger volumes than v3.1 for the juxtacortical, deep white matter, and infratentorial anatomical regions. Using default sensitivity settings, v3.1 quantified a greater number of periventricular lesions, but not a larger volume load. However, using a lower periventricular sensitivity setting of 1.2 vs 1.0 as recommended by Cortechs, periventricular lesion counts were not statistically different for the two versions. A too high sensitivity may give false positive results though; Cortechs has likely kept their default sensitivity setting to avoid false negatives. When using the ML/DL algorithm, whether having no such settings is an advantage needs yet to be confirmed by a radiologist with MS expertise. Our preliminary expert verification and subsequent analysis confirms that the v4.12 segmentations are significantly better classified and filled than the ones of its predecessor. Our findings highlight the enhanced performance of NQ-MS v4.12 driven by the integration of ML/DL, and underscore the significance of improving lesion quantification in MRI analysis. These advancements hold promise for improving the diagnosis and monitoring of neurological conditions. Further research and validation are necessary to explore the full potential of NQ-MS in the clinics.
Wibeke NORDHØY, Yeva PRYSIAZHNIUK, Piotr SOWA, Dragoljub BLAGOJEVIC, Lars SKATTEBØL, Aziz ULUG, Øystein BECH-AASE (Oslo, Norway)
15:30 - 17:00
#47756 - PG281 Tackling the challenge of improving the automated diagnosis of Autism spectrum disorders: assessing the potential of non-standard brain connectivity metrics, deconvolution approaches and machine learning classifiers for the analysis of intrinsic fMRI data.
PG281 Tackling the challenge of improving the automated diagnosis of Autism spectrum disorders: assessing the potential of non-standard brain connectivity metrics, deconvolution approaches and machine learning classifiers for the analysis of intrinsic fMRI data.
Autism spectrum disorders (ASD) are a group of neurodevelopmental conditions characterised by cognitive and/or behavioural impairments. The diagnostic process is multidisciplinary and time-consuming, mainly involving neuropsychological and imaging assessments. Therefore, achieving a fast and precise diagnosis remains challenging. In this context, the use of innovative methodologies for analysing intrinsic functional magnetic resonance imaging data (fMRI), such as non-standard connectivity metrics, deconvolution approaches to mitigate hemodynamic responses, automated diagnosis and biomarker identification of ASD, among others, are promising. The purpose of this work is to evaluate and compare intrinsic fMRI data from subjects diagnosed with ASD and typical controls. To this end, five non-standard functional connectivity metrics, two deconvolution approaches, and seven machine learning classifiers for automated diagnosis were considered.
Raw data were obtained from the publicly available database ABIDE I Preprocessed (Autism Brain Imaging Data Exchange) [1]. Intrinsic fMRI timeseries (AAL atlas, 116 ROIs) [2] from 346 ASD subjects and 369 typical controls were obtained. As shown in Table 1, the following methodologies were applied: six functional connectivity metrics, namely Pearson’s correlation (PC) and five additional non-standard metrics (MULAN toolbox) [3]; two approaches for the deconvolution of the BOLD signal, namely Blind Deconvolution (BD) (MATLAB® rs-HRF toolbox) [4] and Paradigm Free Mapping (PFM) [5]; and seven machine learning classifiers to perform the automated diagnosis of ASD (Malini toolbox) [6]. For each connectivity metric, two different methods were adopted: bivariate, considering only the pairwise connections; and partial, calculating partial results to consider the influence of other ROIs. The connectivity direction was also considered: U and D indicate undirected and directed connectivity, respectively [3] (Figure 1). The following performance metrics were considered for the machine learning classifiers: accuracy, precision, recall/sensitivity, specificity, balanced accuracy and F1-score. The statistical analyses focused on accuracy included: Friedman test to compare the machine learning classifiers with Nemenyi as a post-hoc test; two-way ANOVA to evaluate statistically significant differences in the results of RLR classifier with Bonferroni as a post-hoc test. In this work, 228 different combinations were obtained for the evaluation of the automated diagnosis of ASD using the machine learning classifiers (main results in Table 2). In terms of statistical comparison of the classifiers for accuracy, RLR and LinearSVM demonstrated significantly superior performance, while NaïveBayes and KNN exhibited significantly inferior performance (p<0.001). B computations led to higher accuracies compared to P computations. Regarding the RLR classifier, the metrics BPC, BCorrD, BH2D, BH2U, and BCorrU showed significantly higher accuracies; conversely, the metrics PCorrD-a, PCohW1-a, PCorrU-a, PCohW2-a, and PCorrU-b demonstrated significantly lower accuracies (p<0.001). In relation to the deconvolution approaches, non-deconvolved exhibited significantly superior performance when compared to BD (p=0.0036). As shown in Table 3, regarding the functional connectivity metrics, non-deconvolved demonstrated a superior performance overall, except for TE metric. BD performed better for metrics such as PC, Corr and H2. Conversely, PFM performed better for metrics of the entropy family, such as MIT and TE, as well as PC. Improving the automated diagnosis of ASD using intrinsic fMRI data remains a major challenge due to the disorder’s complexity. In relation to the global accuracy results, the most relevant values achieved presented a performance that is similar to or better than those observed in other studies [7], in particular considering RLR and LDA classifiers. Surprisingly, the highest accuracies were obtained in the non-deconvolved group. In addition, non-standard metrics such as Corr, H2 and MIT demonstrate potential and may be a particularly useful strategy in the search for biomarkers in ASD. A considerable limitation of this work is related to the ABIDE database: although it provides a large number of subjects, which is positive for machine learning computations, it is also heterogeneous, which makes it more difficult to identify ASD biomarkers. For future work, several strategies can be explored, such as optimising parameters of the classifiers already applied, using additional classifiers, and implementing the feature selection approach before the machine learning computations. Improving the automated diagnosis of ASD is particularly challenging; however, several strategies remain to be explored. The highest accuracies obtained in this work are similar to or higher than those observed in other studies. Corr, H2 and MIT demonstrate potential to contribute to the search for biomarkers in ASD.
Lucianna LOPES DO COUTO (Lisbon, Portugal), Sofia REIMÃO, César CABALLERO-GAUDES, Alexandre ANDRADE
15:30 - 17:00
#46467 - PG282 Classification of ica components in complex multi-echo fmri data.
PG282 Classification of ica components in complex multi-echo fmri data.
Independent component analysis (ICA) has become an essential technique for denoising functional magnetic resonance imaging (fMRI) data by decomposing the signal into statistically independent components [1,2]. A critical challenge is accurately distinguishing between components representing blood-oxygen-level-dependent (BOLD) signal and non-BOLD noise components [3,4]. Recently, a new framework for component classification has been proposed for the multi-echo fMRI acquisition, which captures the signal decay across multiple echo times [5,6]. The TE-dependent analysis (TEDANA) pipeline implemented the framework, which derives selected metrics from the multi-echo fMRI data and classifies them using a custom decision tree [7]. This study builds upon TEDANA classification efforts by evaluating multiple classifiers trained on metrics from different representations of complex multi-echo fMRI data, investigating whether incorporating metrics from real and imaginary parts improves classification performance.
The classification dataset was created from manual rating of ICA components by three independent raters, using ICA component metrics derived from the magnitude, real, and imaginary parts of complex multi-echo fMRI data. Metrics included both parameters defined in TEDANA (such as kappa and rho) and custom-made parameters. After removing observations with missing values and components without clear BOLD/non-BOLD classification, the final dataset comprised 998 components: 903 non-BOLD (90.5%) and 95 BOLD (9.5%). We addressed this class imbalance by down-sampling the majority class in the training set to achieve a 1:1 ratio. Variable selection was based on non-parametric correlation analysis. Data were split into training (90%) and testing (10%) sets. Four classifiers were trained and optimized using k-fold cross-validation: Naive Bayes, Logistic Regression, Support Vector Machine (SVM), and Random Forests. Each classifier was trained twice: first using only magnitude-derived metrics, then using all metrics. Classification using only magnitude-derived metrics yielded high test accuracies: Naive Bayes (96.0%), Logistic Regression (97.0%), SVM (92.0%), and Random Forests (94.0%). All classifiers demonstrated high precision for non-BOLD components (98.0%-100.0%) but lower precision for BOLD components (57.0%-82.0%). When trained on all metrics, test accuracies remained similar: Naive Bayes (97.0%), Logistic Regression (93.0%), SVM (89.0%), and Random Forests (91.0%). Only Naive Bayes classifier showed an improvement (+1.0%), while other classifiers showed decreases in accuracy: Logistic Regression (-4.0%), SVM (-3.0%), and Random Forests (-3.0%). Precision for non-BOLD components remained high across all classifiers when using all metrics, but precision for BOLD components decreased for most classifiers compared to using only magnitude-derived metrics. Our findings suggest that incorporating metrics from real and imaginary parts of complex multi-echo fMRI data does not consistently improve the classification of ICA components. While Naive Bayes showed slight improvement with all metrics, other classifiers demonstrated decreased performance. The decreased performance when including all metrics may be attributed to redundant or noisy features complicating decision boundaries, consistent with low correlations between real/imaginary-derived metrics and the outcome variable (mean correlation: 0.178 for real-part, 0.077 for imaginary-part). This study analysed a single train-test split, with variability observed across different training subsets. The high class imbalance remains a challenge despite the down-sampling approach. Future work should include aggregated analyses across multiple train-test splits and feature selection methods to identify the most informative combination of metrics. Overall, magnitude-derived metrics remain most valuable for BOLD vs. non-BOLD classification in multi-echo fMRI data. This study demonstrates that metrics derived from the magnitude representation of complex multi-echo fMRI data provide robust features for classifying ICA components as BOLD or non-BOLD. The overall classification accuracy was 92% or higher. The inclusion of metrics from real and imaginary parts of the data did not yield consistent improvements in classification performance across different algorithms.
The work on this publication was supported by the Czech Science Foundation (GAČR), project No. GA23-06957S.
Jakub JAMARIK (Brno, Czech Republic), Michal MIKL, Martin GAJDOŠ, Radek MAREČEK, Daniel SCHWARZ
15:30 - 17:00
#47870 - PG283 Case study of cohort bias in deep learning-based image segmentation.
PG283 Case study of cohort bias in deep learning-based image segmentation.
At the latest with the publication of the nnU-Net framework [1] and the availability of large cohort studies including magnetic resonance (MR) imaging (e.g., UK Biobank [2] or the German National Cohort, NAKO [3]), large-scale processing of MR images, for example, segmentation of various different target structures (e.g., organs, bones, muscles, or adipose tissue) is widely performed eventually leading to versatile multiclass segmentation models, e.g., TotalSegmentator [4] and related systems [5–7].
The training data sets of these models often show dependencies, i.e., different iterative generations of MR segmentation models are based on output segmentations of former models, eventually leading into a vicious cycle. This could in part be a direct consequence of the time-consuming manual annotation process, usually requiring special training.
Recently, Willem et al. showcased the different biases that can occur along the general machine learning (ML) pipeline [8].
Segmentation performance of (parts of) the human body is directly influenced by its natural variability, besides the primary category of sex along the continuous scales of age and body size (e.g., using body mass index [BMI]). Additionally, important categorical data such as race/ethnicity is seldom collected in a standardized way or underrepresented. It can be assumed that poorly curated training data sets lead to less generalizable segmentation models. Bias introduced by using such data sets is referred to as cohort bias.
Aim of this study was to systematically investigate the influence of cohort bias in the training data set of segmentation models using the example of adipose tissue (AT) segmentation.
3D VIBE Dixon MRI from NAKO (protocoll details to be found in [3]) were used to create 18 distinct training data sets (3 age groups [20–39, 40–59, >60 years], 3 BMI groups [18.5–24.9, 25–29.9, >30 kg/m²] for women/men, respectively) with n=30 each. These sets were used to train 18 distinct nnU-Net models (1 fold, nnU-Net ResEnc M [9], 3d_fullres configuration) [10]. An additional randomly drawn test set from NAKO (n=1000) was used to estimate model performance on the general population. Each model was tested on the 17 remaining biased training data sets and on the test set in terms of segmentation performance (DSC for 2 classes; subcutaneous AT [SAT], visceral AT [VAT]) and AT volumes representing real-world outcomes. Fig. 1 shows the on-average top/bottom 3 models ranked by DSC. On average, the best performing model was trained on data from women between 40 to 59 years with BMI >30 kg/m². By contrast, using data from men <60 years with BMI between 18.5–24.9 kg/m² led to the worst performing models. Fig. 2 summarizes intersectional testing of biased models. On the test set from the general population, mean DSC was 0.977 for SAT and 0.964 for VAT. Volumetric errors range from 0.2–2% (mean 0.7%) for SAT and 0.5–6% (mean 1.8%) for VAT. Best performing model for both classes was trained on overweight women (20–39 years). The composition of the training set affects the performance of the segmentation models. In the present study, n=30 image datasets were used per training dataset. This number has led to very reliable models in the past [10]. It can therefore be assumed that the influence of unbalanced training sets is even stronger in a scenario with limited availability of image datasets, especially in anatomically complex structures, e.g., visceral adipose tissue. Using the example of AT segmentation, it was shown that the composition of the training set according to relevant parameters (in this specific case sex, age and BMI) plays a major role in the development of generalizable segmentation models.
Tobias HAUEISE (Tübingen, Germany), Jürgen MACHANN
15:30 - 17:00
#47112 - PG284 Automatic multi-contrast spinal nerve rootlets segmentation to study spinal-vertebral level correspondence.
PG284 Automatic multi-contrast spinal nerve rootlets segmentation to study spinal-vertebral level correspondence.
MRI images allow identification of spinal nerve rootlets and intervertebral discs (IVDs), which can be used to determine spinal and vertebral levels, respectively [1, 2]. Spinal rootlets are relevant for functional MRI group analyses [3, 4] and neuromodulation therapy [5]; however, their applicability is impacted by considerable variability in spinal and vertebral levels across individuals [2, 6]. A method for automatic rootlet segmentation was proposed [1], but it is limited to a single contrast and dorsal rootlets only. In this work, we developed a model to segment dorsal and ventral C2–T1 rootlets from MP2RAGE and T2w MRI data. Then the model was used to explore the correspondence between spinal and vertebral levels.
Three datasets of healthy adults covering the cervical spinal cord were used for model training: an unpublished dataset of 7T MP2RAGE (3 aligned contrasts: UNIT1, INV1, INV2) 0.7×0.7×0.7 mm data (15 training and 4 testing participants resulting in 45 training and 12 testing images) and two open-access datasets of 3T T2w 0.6×0.6×0.6 to 0.8×0.8×0.8 mm images (spine-generic [7]: 21 training and 3 testing images; and OpenNeuro ds004507 [8]: 10 training and 2 testing images). Ground-truth (GT) labels of dorsal and ventral C2-T1 rootlets were generated semi-automatically for all training and testing images using an existing segmentation method [1] (which segmented only dorsal C2-C8 rootlets), followed by manual correction and annotation of ventral rootlets using FSLeyes viewer.
A multi-contrast segmentation model was developed with the nnUNet framework [9] using 76 training images, trained with 2000 epochs, and 17 testing images with GT masks to compute Dice. To analyse the correspondence between spinal and vertebral levels, we used all the training and testing data and an additional 82 images [7] automatically segmented by the proposed model.
Figure 1 shows the automatic analysis pipeline utilising Spinal Cord Toolbox (SCT) v6.4 [10]. For each image, the spinal cord was segmented [11, 12], IVDs and the pontomedullary junction (PMJ) were identified [13, 14], and nerve rootlets were segmented using the proposed model. Then, IVDs were used to estimate the vertebral levels and rootlets were used to obtain the spinal levels [1]. Lastly, the distance between the PMJ and spinal and vertebral level midpoints was measured along the cord centerline. To account for participants' demographics, we normalised the distances by the participant’s height and multiplied them by the median height (to preserve millimetre units). Figure 2 shows illustrative segmentation by the model for one T2w and one MP2RAGE test image. The model achieved a Dice score of 0.65 ± 0.10 (mean ± standard deviation [SD] across levels and 17 testing images). Figure 3 presents the Dice across levels and contrasts on the testing images. Figure 4a depicts the correspondence between spinal and vertebral levels measured as the distance from the PMJ. Figure 4b shows the Bland-Altman analysis. The bias term for vertebral level C2 (VLC2) and spinal level C3 (SLC3) is zero, suggesting that these levels correspond exactly. A multi-contrast model showed a robust segmentation performance across four contrasts (T2w, INV1, INV2, and UNIT1) and different levels (C2-T1) for both ventral and dorsal rootlets. The analysis of spinal and vertebral level correspondence confirms the previous MRI report [2] and anatomical textbooks [15] that the spinal levels are shifted relative to the vertebral levels and that this shift changes with lower levels. The more caudal the level, the greater the shift (Figure 4a and bias term in Figure 4b across levels), coupled with higher inter-subject variability (more scatter clusters in Figure 4b across levels). In this study, we used the participants' height to account for potential biological differences among individuals. Ideally, the spinal cord or spine length would be considered, but MRI data typically does not cover the entire spine. We developed a deep-learning model for spinal rootlets segmentation across different MRI contrasts and studied the correspondence between spinal and vertebral levels in an adult population. Our results confirm that spinal levels are shifted relative to vertebral levels, and that this shift increases at more caudal levels, accompanied by greater inter-subject variability. Future work will focus on validation using data from participants with spinal pathologies.
Katerina KREJCI (Brno, Czech Republic), Jiri CHMELIK, Sandrine BÉDARD, Falk EIPPERT, Ulrike HORN, Julien COHEN-ADAD, Jan VALOSEK
15:30 - 17:00
#46922 - PG285 Validation of Delta-Radiomics Models to Predict Response to Stereotactic Magnetic Resonance Guided Radiation Therapy of Pancreatic Cancers.
PG285 Validation of Delta-Radiomics Models to Predict Response to Stereotactic Magnetic Resonance Guided Radiation Therapy of Pancreatic Cancers.
For patients with unresectable pancreatic tumors, stereotactic magnetic resonance guided adaptive radiation therapy (SMART) increases survival by delivering a high biologically effective dose in few fractions, although many patients still experience early metastatic or local recurrence [1, 2]. To address this challenge, radiomics leverages mathematical analysis to extract and quantify textural information from medical images, thereby providing additional insights to predict such outcomes. Meanwhile, delta-radiomics assesses the variation in features captured at treatment times and has demonstrated potential in predicting radiotherapy outcomes when combined with machine learning [3]. However, in the case of pancreatic cancer, these studies lack both internal and external validation, which we aim to address here by rigorously showing the performance of such models.
We included all patients treated with SMART for unresectable pancreatic ductal adenocarcinoma from Montpellier Cancer Institute (France) and from University Hospital La Milagrosa, Madrid (Spain). For all these patients, 0.35T T1-w TrueFISP simulation (simu) and fraction (Fn) MRI were acquired. Radiomics features were extracted following IBSI guidelines [4] on the manually annotated gross tumor volume (GTV) ROI. Delta-radiomics features were calculated between different Fn and simu, and reliable features, as identified in a previous study [5], were analyzed. Various feature selection algorithms and predictive machine learning models were evaluated to predict local recurrence at 1 year (LR1y) and metastatic recurrence at 9 months (MR9m) using radiomics or delta-radiomics data from a single time-point. Prediction models were trained on 70% of the Montpellier dataset after feature selection and first evaluated with an internal validation cohort of 25 patients. A subsequent evaluation was performed using an external validation cohort of 37 patients from Madrid, where radiomics features were harmonized using the ComBat method [6]. Both evaluations were performed using bootstrapping (n=200) to compute the 95% confidence intervals for the area under the curve (AUC) and the Brier score. 85 patients were recruited from Montpellier Cancer Institute (France) and 37 from University Hospital La Milagrosa, Madrid (Spain). In the first cohort, 14 presented LR1y and 44 MR9m status while they were 5 and 15 respectively in the second cohort.
In the internal validation group, the Random Forest (RF)+RF model using F3 radiomics data achieved the highest AUC of 0.96 (95% CI: 0.87-1.00) for predicting LR1y, with a Brier score of 0.08 (95% CI: 0.03-0.14), 84% sensitivity, and 100% specificity. For MR9m, the RF+ADABOOST model with F1/F3 delta-radiomics data yielded an AUC of 0.83 (95% CI: 0.66-0.97), a Brier score of 0.22 (95% CI: 0.20-0.24), 91% sensitivity, and 75% specificity.
In the external validation cohort, these models demonstrated an AUC of 0.71 (95% CI: 0.54-0.84) for LR1y and 0.51 (95% CI: 0.28-0.75) for MR9m, with Brier scores of 0.19 (95% CI: 0.11-0.27) and 0.25 (95% CI: 0.22-0.29), respectively. Sensitivity and specificity were 40% and 78% for LR1y, and 40% and 68% for MR9m.
In the external validation cohort, the ANOVA K BEST+PSVM model using Simu radiomics data achieved the highest AUC of 0.80 (95% CI: 0.50-0.99) for LR1y, with a Brier score of 0.25 (95% CI: 0.23-0.27), 80% sensitivity, and 56% specificity. For MR9m, the ANOVA K BEST+RF model using F1/F3 delta-radiomics data achieved an AUC of 0.85 (95% CI: 0.71-0.96), a Brier score of 0.23 (95% CI: 0.22-0.24), 73% sensitivity, and 82% specificity. This study demonstrates the potential of delta-radiomics data combined with machine learning models to predict radiotherapy outcomes for patients with pancreatic tumors. Rigorous internal and external validation was crucial for assessing model reliability and generalizability. While LR1y predictions showed strong performance in internal validation, they significantly declined in the external cohort due to the low incidence of local recurrence, which highlighted the need for recalibration [7]. In contrast, MR9m predictions, although not as strong as LR1y, maintained robust performance with F1/F3 delta-radiomics models. This is particularly encouraging, as metastatic recurrence was the primary cause of mortality in the Montpellier dataset (23 out of 34). Finally, the application of the ComBat method for harmonizing radiomics features was limited by small subgroup sample sizes (n<20), preventing its full utilization. This study highlights the potential of delta-radiomics combined with machine learning to predict radiotherapy outcomes in unresectable pancreatic tumors, demonstrating the importance of rigorous validation. Future work should prioritize the inclusion of additional cases and the enhancement of model calibration. Incorporating clinical and dosimetric features could further augment predictive power and address the challenges observed in the external validation.
Florent TACHENNE (Montpellier), Gladis VALENZUELA, Marion TARDIEU, Olivier RIOU, David AZRIA, Morgan MICHALET, Daniela GONSALVES, Jesus DOMINGUEZ, Felipe COUÑAGO, Stéphanie NOUGARET
15:30 - 17:00
#47864 - PG286 Automatic features extraction for multimodal deep learning fusion of Ultrafast-DCE MRI for breast lesion classification.
PG286 Automatic features extraction for multimodal deep learning fusion of Ultrafast-DCE MRI for breast lesion classification.
Breast cancer is the most common cancer among women worldwide, reaching almost 2.3 million new cases in 2022 [1]. Early detection is critical, and compared to mammography breast magnetic resonance imaging (MRI) shows higher sensitivity, particularly for identifying invasive cancers [2]. Recent advances have focused on ultrafast contrast-enhanced (UF-DCE) MRI protocols, which offer dynamic imaging capturing early enhancement kinetics, enabling potentially better differentiation between benign and malignant lesions [3].
Growing interest has been reported in multimodal AI systems that integrate imaging and non-imaging data to better capture the complexity of patient profiles [4–6], to enhance diagnostic accuracy. Thus automatic multimodal models have been developed to classify breast lesions taking advantage of UF-DCE MRI images, patient data extracted from written reports, and geometric features computed from manual segmentations of lesions [7].
One caveat of these models is the requirement for manual time-consuming segmentation to compute geometric features. Automatic breast lesion segmentation in conventional DCE-MRI, remains challenging due to the low signal-to-noise ratio and significant inter-patient enhancement variability [8]. In this work, we investigate the use of automatic segmentation models in UF-DCE to automatically extract these geometric features.
As shown in Figure 1, the main goal is to remove the need for human intervention in lesion segmentation to derive geometric features, exploited in our multimodal classification approach [7]. To achieve this, we used a custom U-Net model with an automatic extraction processing of geometric features.
While the lesion position is still required to crop around a lesion (volume of interest) due to the absence of a detection model, this step is significantly less time-consuming than full manual segmentation.
Automatic lesion segmentation: a U-Net model [9] was trained using the same images as the one used for the classification model, with a key difference in the cropping strategy. Instead of cropping only around lesions, random regions were sampled within each whole breast, excluding background areas without tissue, increasing the amount of training data for the segmentation model. Lesion position being important for the classification model, particularly in identifying lymph nodes [7], we performed a Global Position Embedding (GPE) by encoding the crop location relative to the whole breast as a three-channel 3D image (one for each dimension) and concatenated it with the 3D last phase UF-DCE subtracted images.
Geometrical features extraction: for each lesion, the U-Net produced a coarse lesion segmentation map that was binarized (background = 0, suspected lesion = 1). Then, we used the SimpleITK library [10] to refine the segmentations by keeping only the region closest to the lesion location and whose size was greater than 50 voxels, as depicted in Figure 2. Using the kept region, geometric features were extracted including bounding box (position and size), volume, center of gravity, ellipsoid axes lengths, elongation, and flatness [7]. If no valid region was found, geometric features could not be extracted and the classification model replaced them with the mean value of each feature. For all experiments, we used a stratified 5-fold cross-validation on a dataset including 987 manually segmented lesions classified as malignant, benign or lymph nodes [7].
On the segmentation task exemplified in Figure 3, when only considering crops centered on lesions, our segmentation model achieved a DICE coefficient of 60.9±1.2%.
On the classification task, we investigated the impact of the automatic features extraction during the training or test stages. We compared two variants, “auto” and “hybrid”, against the reference model [7] which used features from manual segmentations (Figure 4). The auto model used a classifier that was both trained and evaluated on the automatic features, while the hybrid model was trained on the original features but evaluated on the automatic features. Resulting sensitivity, specificity and Area Under the ROC curves (AUC) are reported in Figure 4. Both methods relaying on automatic segmentation underperformed compared to the baseline reference using manual segmentation, which was also reported in the literature [11] . However, the hybrid approach offered a promising compromise between segmentation effort and accuracy. Indeed, this method achieved an AUC only 2.8% lower than the reference, while eliminating the need for manual segmentation at inference time. Additionally, training the classifier on manual segmentations yielded a 3.6% higher AUC than training directly on automatically extracted features. The proposed hybrid method can be used to avoid manually segmenting lesions during clinical practice at a small accuracy cost. Further work should be done to improve the segmentation model and therefore the extracted geometric features.
Valentin DURAND DE GEVIGNEY, Belinda LOKAJ (Geneva, Switzerland), Diogo CORREIA, Luan MURATI, Takeru TSUKADA, Christian LOVIS, Jérôme SCHMID
15:30 - 17:00
#47851 - PG287 Subset Selection in Diffusion MRI: Uncertainty and Degeneracy Assessment with µGUIDE.
PG287 Subset Selection in Diffusion MRI: Uncertainty and Degeneracy Assessment with µGUIDE.
Diffusion MRI enables the estimation of tissue microstructural parameters performing acquisitions with varied acquisition settings. However, the high dimensionality of the acquisition parameter space can result in long acquisition times. Therefore, selecting the most informative subset of measurements to characterize tissue microstructure remains crucial.
Recent work [1] proposed a physics informed machine learning approach that couples a Concrete autoencoder (CA) [2] with a signal forward equation to automatically subselect the most informative dMRI measurements. This approach has shown high accuracy of parameter estimates compared to subsampling based on random, uniform, Cramer-Rao Lower Bound (CRLB) [3] (non-physics informed machine learning strategies).
In this work we further assess uncertainty and degeneracy of biophysical parameter estimation under measurement subset selection by different selection methods using µGUIDE [4], a Bayesian inference framework that estimates the posterior distributions of microstructural parameters using deep learning.
Synthetic data:
The MUDI dataset [5] was used as basis for the training, it consists on a 5D Diffusion-T₁-T₂* protocol acquired on a 3T scanner (80 mT/m) using 1344 unique settings, varying b-value ([0–3000] s/mm²), gradient orientation, inversion time (TI: 20–7322.7 ms) and delay time (TD: [0, 25, 50] ms). The repetition time (TR) was 7500 ms, and the b-tensor anisotropy (bΔ) was 1.
To generate synthetic data for training and evaluation, signals were simulated using the following signal equation:
S = S₀·exp (-b∶ D) · |1 - 2 ·exp(-TI / T₁) + exp(-TR / T₁)| · exp(-TD / T₂*)
And b:D:
b∶ D = (1/3) · b · b_Δ · [D_∥ - D_⊥] - (1/3) · b · [D_∥ + 2 · D_⊥] - b · b_Δ · (g(Θ_g,Φ_g) · g(θ,φ))² · [D_∥ - D_⊥]
Where b is the b-value; bΔ the anisotropy factor of the diffusion encoding; D∥ D⊥ parallel and perpendicular diffusivities; g(Θg,Φg) the direction of the diffusion gradient in spherical coordinates; g(θ,ϕ) the principal direction of the tissue structure; TI, TR and TD inversion time, repetition time and echo delay time, T1 and T2∗ the longitudinal and effective transverse relaxation times.
We used the following ranges to simulate the data:
S_0∈ [0.5,5.0]
T_1∈ [100,5000] ms
T_2^*∈ [0.01,2000] ms
D_∥∈ [0.01,3.2] μm2/ms
D⊥=k⋅D∥D_⊥ = k⋅ D_∥, with k ∈ [0.01,1]
θ∈ [0,π],
ϕ∈ [0,2π]
Synthetic noise was added at an SNR of 30 for the lowest b-value and longest TI/TD.
Measurement subset selection and parameter estimation:
Figure 1 illustrates the proposed workflow. Firstly, measurements were sub-selected using uniform, random, CRLB and physics-informed machine learning selection (CL+eq) (Figure 1 A1); then, the selected measurements were fed to uGUIDE for training and inference of the posterior distribution of microstructural parameters (Figure 1 A2). The selected measurements were fed to uGUIDE (Figure 1 B1). The number of selected measurements were N = 500, 100, 50. Finally, the maximum a posteriori, degeneracy and uncertainty were computed and compared to the full dataset. Results and Discussion:
Figure 2 presents examples of posterior distributions for the estimated parameters. In purple, the posterior distributions obtained using all the signal measurements, it can be seen the full width at half maximum (FWHM) of the distribution is narrow. The posteriors using a decreased number of measurements are shown for all 4 sub-selection methods. As the number of measurements decreases, the posterior distributions remain centered around the ground truth, with a moderate increase in FWHM. This indicates a slight increase in uncertainty, yet the estimates remain stable and well constrained.
Figure 3 presents the Maximum a Posteriori (MAP) estimates for decreasing sample sizes (N=500, 100, 50). The results show that CL+eq consistently yields more accurate MAP estimates with lower uncertainty, particularly at lower sampling rates, aligning with findings of higher accuracy in previous work [1].
Figure 4 presents the uncertainty for the different sub-selection methods. Again, the CL+eq method shows the most consistent dispersion of the most probable samples, indicating robust performance across varying sample sizes. In contrast, Uniform and Random selection strategies show greater uncertainty and variability in the estimates. Although CRLB optimization is grounded in analytical minimization of variance, it is still outperformed by the CL+eq approach.
An exception can be noted for T2∗ , where CL+eq shows reduced performance. This is attributed to the fact that CL+eq was trained on a representative parameter distribution in human data for measurement subset selection, whereas the evaluation here was performed on a uniform distribution including long T2*, and the delay times (TD) included in the acquisition are short. The results reinforce the benefit of physics-informed selection strategies and highlight the value of µGUIDE in assessing the performance of reduced measurement sets.
Maria Paula DEL POPOLO (Utrecht, The Netherlands), Marco PALOMBO, Álvaro PLANCHUELO-GÓMEZ, Maëliss JALLAIS, Chantal TAX
15:30 - 17:00
#47755 - PG288 Unsupervised Clustering Approach for Profiling Multiple Sclerosis White Matter Lesions Through Quantitative and Semi-Quantitative MRI.
PG288 Unsupervised Clustering Approach for Profiling Multiple Sclerosis White Matter Lesions Through Quantitative and Semi-Quantitative MRI.
Multiple sclerosis (MS) is characterized by white matter lesions (WMLs) that exhibit heterogeneous features, reflecting variability across different stages of demyelination, neuroinflammation, and neurodegeneration. Quantitative and semi-quantitative magnetic resonance imaging (qMRI) can help to detect these underlying pathological alterations in vivo.
This study aims to identify different lesion profiles based on qMRI features using an unsupervised machine learning approach.
We included 91 MS patients (76% females; mean age 41.7 ± 13.0 years) who underwent a brain 3T MRI on Philips Elition S scanner at the University Hospital of Verona, Italy. WMLs were manually segmented on T1-weighted (T1w) and T2-weighted (T2w) FLAIR images. Mean qMRI values were extracted from the cores of WMLs with a core volume > 9 mm3.
T1/T2 ratio (T1T2r) was computed from calibrated T1w (T1c) and T2w (T2c) images. Intensity calibration was performed using non-brain tissues (eye and temporalis muscle) [1].
Magnetization transfer (MT) ratio (MTR) was calculated from two 3D GRE acquisitions, with and without MT pulse [2]; MT saturation (MTS) was estimated by integrating an additional T1w image to reduce T1 relaxation time and B1 inhomogeneity effects [3]. T2 relaxation time-based metrics, Myelin, Intra/Extracellular and Free Water Fraction (MWF, IEWF, FWF), Total Water Content (TWC), and T2 of IE water (T2IE), were derived from a multi-echo GRASE sequence using the T2SPARC method [4] for multi-exponential fitting using the toolbox by Canales-Rodríguez (v0.3) [5].
Susceptibility-based metrics included χ-separation and quantitative susceptibility mapping (QSM). χ-separation exploits R2 and R2* (estimated through monoexponential fitting from GRASE and ME-GRE, respectively [6]) to estimate negative χneg (myelin) and positive χpos (iron) sources [7]. QSM was also obtained from the ME-GRE using the QSMxT software toolbox [8]. MRI acquisition parameters are summarized in Tab.1. QSM images were used to identify Paramagnetic Rim Positive and Negative lesions (PRL+/−): PRL+ lesions show a hyperintense rim at the border from iron-laden microglia, while PRL− lesions do not [9].
WMLs qMRI features were scaled using the mean and standard deviation of the normal appearing white matter (NAWM) calculated at a subject level. Principal Component Analysis was applied to the z-scored features for dimensionality reduction, followed by hierarchical clustering using the FactoMineR package (v2.11) [10]. The number of principal components (PCs) to retain was assessed with Horn's Parallel Analysis [11, 12]. The optimal cluster (CL) number was determined using the majority rule among various indices calculated by the NbClust package (v3.0.1) [13]. Differences in qMRI values between WMLs in each CL and NAWM were assessed using Kruskal-Wallis [14] and Dunn’s post-hoc test [15, 16] with Bonferroni correction for multiple comparisons. The effect size (res) was used to evaluate the magnitude of those differences. After clustering, patients were stratified by age quantiles (Q1 to Q4) to examine the distribution of WMLs among CLs. Clustering and statistical analysis were performed in R (v4.4.2) [17]. A total of 720 WMLs were analysed. Four PCs, explaining over 80% of the total variance, were selected. Hierarchical clustering was performed using Euclidean distance and the Ward method and 3 CLs were identified, with an average Silhouette of 0.26. CL1 collected lesion cores with the most altered values compared to NAWM in almost all the MRI metrics (T1T2r, R2, TWC, T2IE, MTS, MTR, FWF, R2*), which similarly contributed to PC1 (R2>0.7, p<0.001) [Fig.1]. CL2 was characterized by the highest values of χpos (res=0.42, p<0.001) and QSM (res=0.45, p<0.001) [Fig.1-2-3], which are the features most correlated to PC2 (R2=0.95 and R2=0.84, respectively, p<0.05). CL3 had less altered WMLs features compared to NAWM (p<0.001). PRL+ had a higher percentage of lesions in CL1 and CL2 (QSM: CL1 56% CL2 35%) with respect to PRL- (CL1 23%; CL2 24%). CL2 lesions number decreased with age (Q1 47.0%, Q4 7.5%), while CL1 and CL3 lesions number increased (Q1 21.9% vs Q4 36.2%; Q1 31.1% vs Q4 56.3%, respectively). Clustering analysis identified 3 distinct WMLs profiles: CL1, characterized by qMRI features indicative of severe tissue disruption more frequently observed in the core of PRL+ lesions; CL2, marked by elevated χpos and QSM values suggestive of iron accumulation in the lesion core consistent with active lesions; CL3, showing milder qMRI alterations relative to NAWM indicative of inactive/remyelinated lesions. The higher prevalence of WMLs in CL2 in younger individuals supports the association of this CL with active lesions, as they seem to decrease with age. Unsupervised clustering based solely on multiparametric qMRI identified distinct profiles of WMLs, reflecting different patterns of tissue alteration and highlighting qMRI's utility in capturing lesion heterogeneity in MS.
Nicola DALL'OSTO (Verona, Italy), Francesco GUARNACCIA, Valentina CAMERA, Laura PASTORE, Rachele BONETTI, Samuele QUAGLIOTTI, Teresa MALTEMPO, Arianna CAVAGNA, Sophia Diana Maria CAMERER, Marco CASTELLARO, Roberta MAGLIOZZI, Francesca Benedetta PIZZINI, Agnese TAMANTI, Massimiliano CALABRESE
15:30 - 17:00
#45658 - PG289 Deep learning accelerated WB MRI protocols for body composition profiling: a pilot study using air coils.
PG289 Deep learning accelerated WB MRI protocols for body composition profiling: a pilot study using air coils.
Magnetic Resonance Imaging (MRI) is a valuable method for detailed body composition analysis, providing detailed quantification of fat and muscle distribution, which is essential for assessing metabolic health [1]. AMRA’s Body Composition Profile (BCP) analysis by segmentation provides clinically relevant data on body fat and muscle composition. It is based on following protocol: six rapid 3D-GRE DIXON imaging stacks, three 14-second LAVA FLEX breath-hold stacks from the neck to pelvis, and three 30-second LAVA FLEX free-breathing stacks from pelvis to knee. This standard protocol, however, relies on the built-in T/R body coil, consequently disabling parallel imaging techniques, thus constraining spatial resolution and/or shorter acquisition times. To address these limitations, our study investigates the development of an advanced BCP protocol incorporating the AIR™ Coil and leveraging the latest MRI advancements, including deep learning-based reconstructions. Validating faster and optimized protocols could streamline MRI-based body composition assessments enhancing efficiency, patient comfort, and clinical utility.
Five healthy volunteers (3 males and 2 females) were scanned using standard BCP protocol with traditional T/R [BODY Coil] as well as three alternative protocols using two 30-Ch AIRTM Coils, along with a built-in posterior array AIR coil: (1) BCP protocol with parallel imaging activation thanks to the use of multi-element coils [AIR Coils] (2) BCP protocol reconstructed with a prototype of AIRTM Recon DL algorithm for DIXON aiming to denoise, remove truncation artifacts in all directions while simultaneously interpolating images [ARDL] and (3) BCP protocol acquired with a new deep learning-based reconstruction algorithm designed for highly undersampled data [DLS]. This algorithm enables increased in-plane spatial resolution keeping similar scan duration. Protocol details can be found in Table 1. For each protocol, DICOM images were transferred to AMRA (Linköping, Sweden), on which body composition analyses were conducted with AMRA® Researcher. Four body composition parameters were obtained for each volunteer: Abdominal Subcutaneous Adipose Tissue volume - ASAT (l), Visceral Adipose Tissue volume - VAT (l), Mean Anterior Thigh Muscle Fat Infiltration - MFI (%), and Total Thigh Fat-free Muscle volume - FFMV (l). To determine whether the alternative accelerated protocols provide results comparable to the standard protocol, the agreement between the standard BCP and the alternative protocols was assessed by calculating the mean differences and the limits of agreement (±1.96*SD). The standard BCP with BODY Coil was considered the reference method, while the BCP with AIR Coils, BCP with ARDL, and BCP with DLS were considered alternative methods. Details can be found in Figure 1. Figures 2 and 3 illustrate typical examples of image segmentation based on the protocols described above.
ASAT: x̄ BODY Coil: 3.85l; AIR Coil, Bias: 0.03, Limits of agreement [-0.10, 0.16]; ARDL, Bias: -0.05, Limits of agreement [-0.21, 0.11]; DLS, Bias: -0.01, Limits of agreement [-0.14, 0.11]; VAT: x̄ BODY Coil: 1.51l; AIR Coil, Bias: 0.01, Limits of agreement [-0.15, 0.17]; ARDL, Bias: 0.02, Limits of agreement [-0.14, 0.17]; DLS, Bias: 0.02, Limits of agreement [-0.12, 0.15]; MFI: x̄ BODY Coil: 4.10%; AIR Coils, Bias: -0.01, Limits of agreement [-0.18, 0.16]; ARDL, Bias: -0.49, Limits of agreement [-0.75, -0.23]; DLS, Bias: -1.16, Limits of agreement [-1.33, -0.99]; FFMV: x̄ BODY Coil: 11.79l; AIR Coils: Bias: -0.05, Limits of agreement [-0.18, 0.09]; ARDL: Bias: -0.13, Limits of agreement [-0.22, -0.03]; DLS: Bias: -0.03, Limits of agreement [-0.19, 0.12]. Analysis of the results indicates that all alternative protocols show a slight bias relative to the standard BCP protocol using T/R BODY coil. BCP with Air Coils and parallel imaging activation demonstrated the closest agreement across ASAT, VAT, MFI and FFMV measurements, with minimal bias and narrow limits of agreement, suggesting it may be a reliable alternative. The BCP with DLS aligned well with the standard BCP but showed slight variability, particularly in MFI measurements – possibly due to the higher resolution. ARDL algorithm, while generally consistent, tended to underestimate values for FFMV. Overall, the variability between protocols was small and comparable to intra-scanner test-retest variability of the standard BCP protocol, except for MFI where the variability was comparable to inter-scanner variability [2]. Applying AIR Coils shows the best agreement with the standard BCP, offering a faster, reliable option for body composition profiling. The BCP with DLS or ARDL provide a much faster viable alternative with slight limitations at this moment. These findings suggest AIR Coils with DL reconstruction could replace the BCP standard, improving efficiency, comfort, possibly extendable to whole body MRI applications, by dramatically reducing scan time.
Manon ROOSE (Jette, Belgium), Julie POUJOL, Arnaud GUIDON, André AHLGREN, Maarten NAEYAERT, Peter VAN SCHUERBEEK, Hubert RAEYMAEKERS
15:30 - 17:00
#47118 - PG290 Automatic method for detection of coronary artery stationary periods using a time-series prediction deep-learning model.
PG290 Automatic method for detection of coronary artery stationary periods using a time-series prediction deep-learning model.
Coronary magnetic resonance angiography (MRA) is a technique used to obtain morphological information about coronary arteries by collecting data during their stationary periods. The stationary period of coronary arteries is visually determined by an operator using cine images; however, this method is time-consuming and operator-dependent. Therefore, attempts have been made to automate the detection of stationary periods. Automatic detection of the stationary period of coronary arteries consists of two steps. In Step 1, the position of the coronary arteries is detected, and a motion curve is generated based on the positional changes across frames. In Step 2, the stationary period is obtained from the motion curve. In a previous study proposing a method using deep learning, the stationary period was determined using a threshold method after the position of the coronary artery was detected using deep learning [1, 2].
We previously proposed an automatic technique that uses deep learning for position detection and stationary period determination [3]. It uses four-chamber cine images to detect the positions of the coronary arteries using a single-shot multibox detector (SSD), a type of convolutional neural network, and then performs image segmentation using U-Net on two-dimensional regions composed of the motion curves of the left and right coronary arteries. In this study, we investigated whether it is possible to automatically determine the stationary period using a time-series prediction deep-learning model for cardiac cine images, which are essentially time-series data.
Cine images of 31 to 91 phases were acquired using 1.5-T and 3.0-T magnetic resonance imaging systems, and the positions of the coronary arteries were detected using SSD for a total of 1,031 cine images. Next, the motion curve of the coronary artery was calculated from the change in the position of the coronary artery between frames and used as the training and test data. The deep-learning neural networks used to detect the stationary period include long short-term memory (LSTM), gated recurrent unit (GRU), and transformer, which are representative models for time-series data. The evaluation of stationary period detection was conducted using recall, specificity, and their geometric mean (G-Mean). The timing of the start and end of the stationary period was represented by the phase number of the cine image, and the detection error (in phases) was obtained by calculating the mean squared error between the detected stationary period and the stationary period (label) visually evaluated by an experienced operator. The detection error (in phases) was calculated and evaluated at eight points, four points each on the left and right, namely, the start and end of the systole and diastole of the coronary artery. The computer used had 32GB of memory, Core i7 13620H CPU (Intel Corporation), an NVIDIA RTX-4070 GPU, an ADAM optimizer, and a learning rate of 0.001. Training was performed until the minimum loss was achieved, and the number of epochs was 8600 for GRU, 10,550 for LSTM, and 20,000 for transformer. The results for GRU, LSTM, and transformer were 0.93, 0.93, and 1.00 for recall, 0.87, 1.00, and 0.99 for specificity, and 0.90, 0.96, and 0.99 for G-Mean, respectively (Table 1). The detection error in mean squared phase errors was 2.17 for GRU, 0.99 for LSTM, and 0.70 for transformer. Except for specificity, which was almost the same as LSTM, transformer showed the best results in all other indices (Fig 1-3, Table 1). Between GRU and LSTM, the latter showed better results. GRU has a simpler gate structure than LSTM does; therefore, the computational load is lighter. However, LSTM is thought to be superior in terms of long-term memory retention. This might have been the reason for LSTM performing better than GRU in terms of memory of the various coronary artery movements associated with heartbeat, including individual differences. However, transformer showed the best results in all other indices except for specificity, compared with GRU and LSTM. Unlike GRU and LSTM, transformer does not use a sequential process but uses an “attention mechanism” that focuses on all time-series data at once. Therefore, transformer can efficiently focus on the positions of the stationary period in the entire cardiac phase, which is thought to enable a more stable and accurate detection of stationary periods. However, because the results of this study were based on a relatively small amount of data, further studies with larger amounts of data are required. In this study, the deep-learning time-series prediction models LSTM, GRU, and transformer, which handle time-series data, were demonstrated to be capable of estimating coronary artery stationary periods. In particular, transformer demonstrated high accuracy in detecting the stationary period of the coronary arteries and is expected to be useful in improving image quality and reproducibility of coronary MRA.
Sanae TAKAHASHI (Tokyo, Japan), Shigehide KUHARA, Yuta ENDO, Haruna SHIBO, Takeyuki HASHIMOTO, Junichi TAKEUCHI, Kenichi YOKOYAMA
15:30 - 17:00
#46156 - PG291 Comparison of manual vs. Artificial Intelligence-based muscle segmentation for evaluating disease progression in patients with CMT1A.
PG291 Comparison of manual vs. Artificial Intelligence-based muscle segmentation for evaluating disease progression in patients with CMT1A.
Intramuscular fat fraction (FF), assessed with quantitative MRI (qMRI), has emerged as one ofthe few responsive outcome measures in CMT1A patients. The main limitation for its use in futuretrials is the time required for the manual segmentation of individual muscles. This study aimed atassessing the accuracy and responsiveness of a fully automatic Artificial Intelligence-based (AI)segmentation pipeline to evaluate disease progression in a cohort of CMT1A patients.
Twenty CMT1A patients were included in this observational prospective longitudinal study. FF was measured twice a year using qMRI in the lower limbs.
CMT1A patients were positioned supine while the leg and thigh of the non-dominant limb were imaged using a combination of flexible coils on the top and spine coils integrated into the scanner bed on the bottom. After a localizer, sets of images were recorded in the axial plane at 1.5T (MAGNETOM Avanto, Siemens Healthineers, Erlangen, Germany) during approximately 45 minutes. Anatomical and quantitative imaging sequences were used in order to compute Fat Fraction (FF) through the generation of quantitative FF maps. The same protocol was conducted at 12 months for the follow-up evaluation (Figure 1).
Muscle segmentation was performed fully automatically using a trained convolutional neural network with or without a human quality check(QC). The corresponding results were compared with the Dice similarity coefficient (DSC) with those obtained from a fully manual (FM) segmentation. In each case, FF progression and its standardized Response Mean (SRM) was computed in individual muscles over the single central slice and a 3D muscle volume at thigh and leg levels to define the most sensitive region of interest. The technical performances and limitations of manual segmentation were assessed through an initial analysis of the between- and within-operator variations and so in a sample of ten CMT1A thigh level images from our cohort. The corresponding DSC values between the two operators were very high (0.92 ± 0.02) (Figure 2).
AI-based segmentation showed excellent DSC values (>0.90). A significant global FF progression was observed at thigh (+0.71 ± 1.28%; p=0.016) and leg levels (+1.73 ± 2.88%, p=0.007), similarly to what was computed from the FM technique (p=0.363 and p=0.634). FF progression of each individual muscle was comparable when computed either from the central slice or the 3D volume.
In order to identify the most sensitive region of interest (ROI), FF progression rates together with the SRM values were assessed using AI-based segmentations methods in a single central slice and within the whole 3D volume. The optimal SRM value (0.70) was actually obtained for the global leg 3D volume analysis using the AI+QC segmentation method.
The time necessary for the fully automatic segmentation process using AI with a QC was10 hours for the entire dataset as compared to 90 hours for the FM. We initially appraised the performance of the CNN-based automatic segmentation process considering the manual segmentation technique as the ground truth. The accuracy of the AI-based segmentation was excellent as illustrated by the >0.90 DSC value obtained for all analyzed muscles. This excellent DSC value was similar to what we previously described and slightly better than the values reported in the literature with other CNN. These values should be definitely analyzed in view of the limitations of the manual segmentation process.
The absence of any significant difference in our study between the FM and the AI-based segmentation techniques regarding the FF progression rate over one year supports that qMRI coupled with AI-based segmentation techniques could be used in future longitudinal studies in CMT1A patients. As such, slowly progressive disease such as CMT1A could be followed over short periods of time on the basis of FF changes computed using qMRI and AI-based segmentation.
Given the slow FF progression at thigh level and the large heterogeneity between muscles and individuals, FF should be quantified from a 3D volume at the leg level for longitudinal analyses. In conclusion, AI-based segmentation technique applied to neuromuscular qMRI is ready to be used in longitudinal studies to assess disease severity and progression in CMT1A patients. Of interest, such an approach makes it possible a 3D volume and individual muscle analysis in the lower limbs. The addition of a QC step is still recommended as it might further improve the FF responsiveness in follow-up studies with higher SRM.
Etienne FORTANIER (Marseille), Marc-Adrien HOSTIN, Constance P MICHEL, Emilien DELMONT, Maxime GUYE, Marc-Emmanuel BELLEMARE, Shahram ATTARIAN, David BENDAHAN
15:30 - 17:00
#47638 - PG292 Combining multi-view object detection for improved brain tumor detection applied to flair mr images.
PG292 Combining multi-view object detection for improved brain tumor detection applied to flair mr images.
Gliomas are a common primary brain tumor, originating from glial cells, and are associated with poor prognosis, particularly in high-grade forms like glioblastoma [1]. Magnetic Resonance Imaging (MRI) is crucial for diagnosis, offering detailed structural images without ionizing radiation. However, manual segmentation of MRI scans is time-consuming and prone to inter-observer variability, affecting diagnostic accuracy [2]. Deep learning models, such as YOLO, have been applied for automated tumor detection in medical imaging, with YOLOv11 [3] demonstrating improved speed and accuracy. Despite these advancements, many models analyze MRI slices independently, overlooking the three-dimensional context of the brain. To address this, we propose a novel pipeline using YOLOv11 with a 2D+ approach, incorporating data from two orthogonal planes, comparing different combinations for axial, sagittal, and coronal. Our hypothesis is that this method will improve tumor localization by combining 2D anatomical views while maintaining computational efficiency.
Our goal is to enhance clinical workflows, reduce manual annotation, and enable faster, more personalized treatment for glioma patients.
The workflow includes data processing, label generation, training, evaluation, and combining 2D labels for 3D masking. We used the BraTS2021 dataset [4], selecting the FLAIR modality for its sensitivity to edema around tumor cores. The 3D MRI volumes were converted into 2D JPEG slices, each paired with a segmentation mask for tumor identification. Bounding boxes were automatically computed from the ground truth segmentation masks. Maximum intensity projections in axial, sagittal, and coronal planes were used to derive 2D bounding boxes, and the final 2D+ bounding box was created by combining them. Labels in YOLO format were generated, and YOLOv11 was trained with an image resolution of 640×640 pixels. The model was tested on 2D slices, and predictions were mapped onto 2D+ space.
Combined binary masks were used to crop the MRI volume, resulting in a tumor-focused sub-volume saved in NIfTI format. Model performance was evaluated using standard metrics such as precision, recall, and mean average precision (mAP) [5]. The overlap between predicted 2D+ masks and ground truth was quantified using the Dice Coefficient, demonstrating the effectiveness of the 2D+ tumor localization approach. The YOLOv11 model trained on the 2D FLAIR MRI slices from the BraTS2021 dataset achieved effective tumor localization across the three anatomical planes. Quantitatively, the model obtained a mean precision and recall of 0.926 and 0.790 on the test set, reflecting the model’s ability to correctly identify tumor regions with a few false positives. After focusing on the 2D space detections, the combined 2D+ segmentation masks were compared against the ground truth. The resulting 2D+ masks enabled improved performance compared to individual 2D models, with the best results being obtained by combining Coronal and Axial views with a Precision of 100% and a Recall of 97%. This study demonstrates that the YOLOv11 2D+ pipeline is a highly effective approach for brain tumor detection in MRI scans. The model achieved high mean precision, recall,
and mAP values across the test set, indicating strong detection accuracy and robustness. Importantly, it maintained consistent performance across various tumor sizes and across the three MRI planes: axial, sagittal, and coronal. A major advantage of the YOLOv11 framework is its real-time inference capability, which, combined with its scalability to large datasets, makes it suitable for clinical environments where speed and efficiency are critical. The low rate of false negatives further enhances its
potential utility in reducing diagnostic errors and radiologist workload.
In addition to detection, the fusion of multi-view 2D predictions enables the generation of precise tumor-centered 2D+ sub-volumes. These focused regions improve the efficiency of downstream tasks, such as radiomics analysis, segmentation refinement, and treatment planning, by eliminating the need to process the entire MRI images [6].
The integration of this pipeline into clinical workflows could provide valuable support in neuro-oncology, particularly for early tumor diagnosis and intervention planning. The YOLOv11 2D+ pipeline delivers accurate and efficient brain tumor detection across multiple MRI planes. Achieving a Dice Coefficient of 0.892 with minimal false positives,
the model enables the generation of focused tumor sub-volumes that streamline downstream analysis. These results highlight the potential of YOLOv11 as a practical tool to support radiologists and enhance early diagnosis in clinical neuro-oncology workflows.
Catarina SANTOS PALMEIRÃO (Lisbon, Portugal), Catarina PASSARINHO, Ana MATOSO, Marta P. LOUREIRO, José Maria MOREIRA, Patrícia FIGUEIREDO, Rita G. NUNES
15:30 - 17:00
#47643 - PG293 Integrative multi-omics modeling enhances Alzheimer’s disease prediction through hybrid deep learning and interpretable machine learning approaches.
PG293 Integrative multi-omics modeling enhances Alzheimer’s disease prediction through hybrid deep learning and interpretable machine learning approaches.
Alzheimer's disease (AD) is a multifactorial neurodegenerative disorder lacking effective curative therapies. Increasing availability of high-dimensional biomedical data has facilitated integrative strategies for early diagnosis and risk prediction. This study aims to develop and validate an integrative predictive modeling framework for AD by combining neuroimaging, genomic, and proteomic data using advanced machine learning and deep learning methodologies.
We analyzed data from the Alzheimer’s Disease Neuroimaging Initiative (ADNI), involving 469 participants classified into AD (n=153), mild cognitive impairment (MCI; n=157), and normal controls (NC; n=159). The dataset included structural magnetic resonance imaging (3D-T1 MRI), genome-wide single nucleotide polymorphism (SNP) genotypes, and plasma proteomic profiles. Radiomic features were derived through contourlet-wavelet transformations combined with gray-level co-occurrence matrix metrics. Genomic and proteomic features underwent dimension reduction via adaptive elastic net and group least absolute shrinkage and selection operator (LASSO) methodologies. Multiple classifiers—support vector machines (SVM), random forest (RF), gradient boosting machines (GBM), and extreme gradient boosting (XGBoost)—were compared. Additionally, a three-dimensional convolutional neural network (3D-CNN) was trained on voxel-level imaging data, with a hybrid 3D-CNN-SVM architecture subsequently constructed for enhanced diagnostic accuracy. Model performance was rigorously evaluated using 10-fold cross-validation and confirmed on an independent validation set, measuring accuracy, sensitivity, specificity, and area under the receiver operating characteristic curve (AUC). Model interpretability was assessed using SHapley Additive exPlanations (SHAP) and root mean square error (RMSE) analyses. The hybrid 3D-CNN-SVM model exhibited superior performance in three-class diagnostic classification (NC vs. MCI vs. AD), achieving an accuracy of 92.11% ± 2.31%, significantly surpassing 2D-CNN and standalone machine learning models (P < 0.05). In binary classification tasks, the model achieved near-perfect accuracy (99.1%) and an AUC of 1.000 for discriminating AD from NC subjects. XGBoost classifiers built separately on genomic, proteomic, and integrated multi-omics data yielded accuracies of 92.9%, 98.8%, and 92.1%, respectively. Notably, the multi-omics XGBoost model augmented with demographic covariates (age, sex, education) provided optimal predictive performance for MCI-to-AD conversion (AUC: 0.945 [95% CI: 0.917–0.982], sensitivity: 0.976, specificity: 0.930). SHAP analyses identified critical protective biomarkers (e.g., rs7988039, apolipoprotein A-II) and risk-associated features (e.g., rs7191801, thrombospondin levels), correlating significantly with disease progression. Our findings highlight the improved diagnostic accuracy afforded by integrative multi-omics modeling over single-modality approaches. The hybrid deep learning framework (3D-CNN-SVM) effectively captures neuroanatomical spatial patterns, while feature-informed XGBoost models capitalize on complementary information inherent to different omics data. Importantly, SHAP-driven interpretations provided biologically meaningful insights into AD pathogenesis, underscoring the clinical translational potential for personalized risk stratification. This study establishes a robust, reproducible, and interpretable computational pipeline leveraging multi-omics integration for precise AD risk prediction. The synergistic combination of deep learning and interpretable machine learning with omics-derived biomarkers presents substantial promise for precision diagnosis and targeted early interventions in high-risk populations.
Runhuang YANG (Beijing, China), Siqi YU, Zhiyuan WU, Haiping ZHANG, Haibin LI, Xiuhua GUO
15:30 - 17:00
#46112 - PG294 Unsupervised clustering of high-field MRI radiomic maps for ovarian cancer tissue characterization.
PG294 Unsupervised clustering of high-field MRI radiomic maps for ovarian cancer tissue characterization.
Ovarian cancer is a biologically heterogeneous disease, frequently diagnosed at an advanced stage with peritoneal dissemination. Identifying tumor compartments prior to histological analysis remains a major challenge, particularly for guiding biopsy and treatment planning [1]. Voxel-wise radiomics applied to high-field ex vivo MRI offers an opportunity to map tumor heterogeneity at high resolution [2]. We present preliminary results of habitat-based unsupervised clustering applied to texture-based radiomic maps derived from 9.4 T preclinical MRI of fixed ovarian tumor and peritoneal carcinomatosis samples.
Five ex vivo tumor samples were collected from two patients, including both primary ovarian tumors and peritoneal metastases. Samples were fixed in formalin for 48 hours and imaged in Fluorinert on a 9.4 T preclinical MRI scanner (fat-saturated gradient echo sequence, TR = 2000 ms, TE = 9.14 ms, flip angle = 60°, 4 averages). MRI preprocessing included N4 bias field correction, Gaussian filtering, and gray-level discretization (bin width = 25). Voxel-wise GLCM features were extracted using a 3×3×3 sliding window centered on each voxel within a segmentation mask [3].
To reduce feature redundancy and retain only informative descriptors of tumor heterogeneity, empirical entropy was computed within the segmentation mask for each GLCM feature. The most spatially diverse and non-redundant features were then selected for clustering. A KMeans clustering was then performed on the voxel-wise parametric maps to delineate distinct imaging habitats. Habitat maps were visually assessed and compared to digitized histological sections, which were aligned to the MRI acquisition plane and segmented using QuPath based on expert annotations. Based on the entropy analysis, four GLCM features were retained for clustering: Difference Entropy, Correlation, Inverse Difference (Id), and Inverse Variance (Figure1)
Unsupervised clustering was initially performed with KMeans (K=2), but this configuration consistently yielded a marginal habitat confined to the edges of the segmentation mask, likely reflecting an edge effect introduced by the radiomic sliding window. To overcome this, clustering was repeated with K=3, which revealed two spatially coherent internal habitats and a third peripheral one. This marginal habitat was then merged with its adjacent internal zone based on spatial continuity, resulting in a final binary habitat segmentation. The two resulting radiomic habitats showed visual strong correspondence with histological compartments across all five samples, with one region aligning with densely cellular tumor areas and the other with stromal or low-cellularity regions (Figure 2). This work demonstrates that unsupervised clustering of voxel-wise GLCM features from high-field (9.4 T preclinical) ex vivo MRI can delineate distinct imaging habitats within ovarian cancer tissue. Entropy-based feature selection enabled the identification of spatially diverse and complementary texture parameters. These features captured complementary textural information, reflecting both heterogeneity (Difference Entropy), structural organization (Correlation), and local uniformity (Id and Inverse Variance).
After correcting for edge-related artifacts, the resulting radiomic habitats exhibited reproducible spatial organization and strong concordance with histological compartments across all samples.
The next step is to spatially register and overlay these MRI-derived imaging habitats with QuPath-based histological segmentations. This alignment will enable voxel-level tissue classification and support the development of supervised models for automated tissue-type prediction. These preliminary results suggest that radiomic clustering based on GLCM features from high-field ex vivo MRI can reveal biologically meaningful imaging habitats in ovarian cancer tissue. Future work will extend this approach to quantitative MRI sequences (such as T1 and T2 mapping), integrate spatial transcriptomics, and develop predictive models toward robust radiogenomic signatures for comprehensive tissue characterization.
Margaux VERDIER (Montpellier), Marion TARDIEU, Maïda CARDOSO, Lakhdar KHELLAF, Pierre-Emmanuel COLOMBO, Christophe GOZE-BAC, Stephanie NOUGARET
15:30 - 17:00
#47311 - PG295 Addressing bias in FreeSurfer imaging-derived phenotypes arising from T2-FLAIR scan omission in UK Biobank using harmonization approaches.
PG295 Addressing bias in FreeSurfer imaging-derived phenotypes arising from T2-FLAIR scan omission in UK Biobank using harmonization approaches.
The UK Biobank (UKB) is collecting multimodal brain MRI scans of 100,000 participants using the same acquisition protocol on identical scanners and extracting imaging derived phenotypes (IDPs) with a standardised pipeline [1-2]. However, confound effects are still present and need to be carefully addressed. In previous work [3], it has been shown that when the T2-FLAIR image is absent, FreeSurfer [4-5] IDPs are affected.
While only around 2% of the participants in UKB are missing T2-FLAIR images, this accounts for up to ~14% variance explained and ~7% of unique variance explained in some IDPs [3], which is substantial enough to cause significant confounding when investigating associations with non-imaging variables (nIDPs). In this work we sought to investigate the prevalence and size of this T2-FLAIR “batch effect” confound across structural IDPs. We then assessed strategies for mitigating this effect, comparing established harmonization approaches. Finally, we investigate the impact of not correctly addressing this bias in downstream analysis on the associations between affected IDPs and non-imaging variables (nIDPs).
We first looked at the magnitude of the T2-FLAIR batch effect across 1432 structural IDPs, using Cohen’s d and examining the ratio of variance between the two groups (T2-FLAIR available vs no T2-FLAIR). We also created age nomograms for IDPs that are known to change with age to show potential downstream effects not addressing this bias.
We then compared several methods addressing this bias while also handling four other standard confounds (age, sex, head position and head size scaling). We first used multiple linear regression of the four confounds and a binary indicator for the missing T2-FLAIR. We then used a modified form of ComBat, which instead of preserving the other confound variables, uses the predicted coefficients to remove their effects from the data at the point at which we apply the batch correction [6-7]. Additionally, to test Combat's advantages over regression, we tested our modified method in two ways: (A) turning off ComBat’s batch rescaling of the IDPs (leaving just the mean shift or bias correction); (B) modifying ComBat to only pool prior distributions for variance scaling and mean shift within classes of IDPs, and not across all different classes (e.g., cortical thickness vs surface area). We validated the effectiveness of these approaches against each other and for when no correction had been applied by comparing Cohen’s d and within-group variances. Finally, we assessed some potential downstream analysis implications of not addressing this effect by computing the Fisher-transformed correlation coefficients with a set of over 18,000 nIDPs, comparing our different test cases against each other and against the case where no correction for missing T2-FLAIR had been applied, using Bland-Altman plots. We found a strong negative effect size between the two groups in over 250 cortical volume and thickness IDPs. In the most affected IDPs, the ratio of variance between the groups with and without T2-FLAIR was as high as 2.8, with the difference being most prominent in FreeSurfer cortical thickness IDPs (Figure 1). When using ComBat, the effect size (mean shift bias) is reduced to below 0.1 for all IDPs, the variance ratio is closer to 1 and the mean shift across ages is corrected (Figure 2 and 3). When using simple regression, the effect size is approximately zero for all IDPs, but the ratio of variance is uncorrected. When looking at associations with nIDPs, ComBat gives stronger correlations than no handling of missing T2-FLAIR or handling via simple regression (Figure 4). When comparing ComBat pooling its priors only within IDP groups, and without the scaling term, we found that ComBat run on all IDPs at once and with the variance scaling term included showed the best reduction of bias shift, a variance ratio closest to 1, and marginally stronger associations with nIDPs. The omission of T2-FLAIR causes a strong bias in the mean and variances of over 250 IDPs. We have shown ComBats advantage over simple regression in its ability to adjust the variance of the data. Through comparisons of the correlations with nIDPs, we have shown that using ComBat to handle both confound and batch correction is beneficial when compared to simple regression. Despite different groups of FreeSurfer IDPs not being affected in the same way by the omission of T2-FLAIR, pooling ComBat priors from all IDPs seems to be beneficial. Additionally, when the bias affects more than just the means, the scaling correction is a beneficial addition over simple regression. To conclude, treating the missing T2-FLAIR effect as a confound and addressing it using regression doesn’t sufficiently correct for its effects. Researchers using FreeSurfer derived IDPs for their own data, or using UK Biobank IDPs should be aware of this and consider using ComBat to handle both the T2-FLAIR effect and other confound corrections.
Jacob TURNBULL (Oxford, United Kingdom), Gaurav BHALERAO, Fidel ALFARO-ALMAGRO, Stephen SMITH, Ludovica GRIFFANTI
15:30 - 17:00
#47842 - PG296 Test-retest reliability of resting-state fMRI functional connectivity: impact of scan length and sample size.
PG296 Test-retest reliability of resting-state fMRI functional connectivity: impact of scan length and sample size.
The consistency of fMRI measurements upon repetition, known as test-retest reliability, is essential for interpreting the validity of neuroimaging results, as measures must be reliable to have statistical power. However, quantifying the reliability of resting-state fMRI is complex. Previous research has indicated poor reliability for univariate functional connectivity measures at the edge level [1]. In contrast, multivariate metrics reflecting whole-brain patterns tend to demonstrate greater reliability [2]. Additionally, the duration of fMRI scans has been found to influence reliability estimates [3]. Despite significant efforts to evaluate test-retest reliability in resting-state fMRI, a comprehensive comparison of various reliability metrics across intra- and inter-session data, while systematically varying scan lengths and sample sizes, is still required. We aim to investigate the intra- and inter-session test-retest reliability of both univariate and multivariate resting-state fMRI functional connectivity metrics under different experimental conditions.
We analysed minimally pre-processed resting-state fMRI data from 100 unrelated healthy adults (54 females, 46 males, ages 22–35) provided by the Human Connectome Project. Data included two sessions (REST1 and REST2) and two runs within the first session (REST1_LR and REST1_RL). Following high-pass filtering and spatial smoothing using FSL, we assessed test-retest reliability both within and between sessions. We systematically varied the number of volumes (300, 600, 900, 1200) and sample sizes (10, 20, 50, 100). Both a voxel-based and a node-based analysis were conducted: 1) Group Independent Component Analysis (ICA) with 50 components was conducted to identify canonical resting-state networks (RSNs), and the spatial similarity between these ICA-derived networks and template RSNs was quantified using the Dice coefficient [4]; and 2) Functional connectivity matrices were computed using Pearson correlation on data that was parcellated into 400 regions based on the Schaefer atlas. We evaluated one univariate metric, the average intraclass correlation coefficient (ICC) across all edges within each RSN [5], as well as two multivariate metrics: fingerprinting, which compares the correlation between connectomes [6], and discriminability, which evaluates the distance between connectomes [7]. At the group level, the spatial similarity between ICA-derived networks and canonical RSN templates remained consistent across all seven RSNs, regardless of the number of volumes analysed (Figure 1A). Spatial reproducibility of ICA-derived networks, when comparing runs within and between sessions, varied by RSN (Figure 2A). The Visual Network (VN), Sensorimotor Network (SMN), and Frontoparietal Network (FPN) demonstrated the most consistent Dice coefficients across different scanning lengths, particularly when considering 50 and 100 subjects.
At the individual subject level, mean spatial similarity increased with longer scan durations and remained relatively stable across varying numbers of subjects (Figure 1B). Similarly, spatial reproducibility within and between sessions improved with increasing scan length, while the number of subjects had a less significant impact (Figure 2B).
For connectivity matrix-based metrics, edge-level ICC reliability improved with longer scanning durations, transitioning from poor to fair reliability (ICC > 0.4) (Figure 3A). The FPN, SMN, Dorsal Attention Network (DAN), and VN generally showed the highest edge-level ICC values. Additionally, the multivariate metrics, differential identifiability and discriminability, both increased with longer scanning lengths (Figure 3B). This study demonstrates the impact of scan length and sample size on the test-retest reliability of various fMRI metrics. While group-level ICA for network identification remains robust across scan lengths, the reliability at the individual level improves with longer scan durations.
Edge-level functional connectivity shows modest improvements with extending scanning times. In contrast, multivariate whole-brain metrics demonstrate greater reliability with increased scan length, suggesting that whole-brain patterns tend to be more stable than individual connections. We show strong variations of the test-retest reliability across various resting-state fMRI functional connectivity metrics, across scanning durations and sample sizes.
Beatriz VALE (Lisbon, Portugal), Marta CORREIA, Patrícia FIGUEIREDO
15:30 - 17:00
#46043 - PG297 Non-invasive Microstructural Characterization of Iron Overload in Aceruloplasminemia Using Static Dephasing Theory.
PG297 Non-invasive Microstructural Characterization of Iron Overload in Aceruloplasminemia Using Static Dephasing Theory.
Aceruloplasminemia (ACP) is a rare neurodegenerative disorder characterized by a complete loss of ceruloplasmin ferroxidase activity, leading to systemic iron overload in the brain, liver, and pancreas [1]. While MRI-based iron mapping, using R2* or Quantitative Susceptibility Mapping (QSM) is widely applied, microstructural iron quantification faces unresolved challenges due to the multiple susceptibility sources which contribute to the measured MR signal [2]. Microstructural iron quantification using MRI microscopy images with a resolution in the µm scale has been extensively studied, however reconstructing magnetic contributions from images in the mm scale, as acquired in a clinical setting, remains complex with inherent mathematical limitations in certain regimes [3].
This study leverages the Static Dephasing Theory (SDT) [4], a framework modeling transverse relaxation, to quantify iron accumulation in ACP brain tissue. SDT describes relaxation in the presence of magnetized objects (e.g. iron-rich cells, blood vessels) that induce local magnetic field inhomogeneities. The theory applies in the regime where these magnetic perturbers are sufficiently large, such that diffusion has negligible effect on the relaxation process. We demonstrate SDT’s capacity to extract microstructural biomarkers (e.g. inclusion volume fractions, absolute magnetic susceptibility) in ACP, advancing beyond conventional R2* or QSM.
One patient with ACP and a matched healthy control were imaged on a 3T MRI system (MAGNETOM Skyra, Siemens Healthineers, Erlangen, Germany). The following sequences were acquired: T1-weighted MPRAGE for anatomical segmentation, multi-echo GRE for QSM and R2* mapping, and dual-echo SPACE for R2 mapping. T1-weighted images were registered to GRE space using FLIRT, and segmentation was performed with FSL FIRST. R2* maps were computed using ARLO, and QSM reconstruction included ROMEO phase unwrapping [5], V-SHARP background field removal [6], and iLSQR dipole inversion [6], with susceptibility values referenced to the CSF. Using SDT, we modeled the signal decay to extract additional microstructural parameters, such as volume fraction and absolute susceptibility of magnetic inclusions, which are not accessible with standard approaches. The static dephasing signal was numerically evaluated and fitted to a voxel subset in the basal ganglia using the L-BFGS-B algorithm with biophysical motivated constraints on the microstructural parameters. Figure 1 provides an overview of the R2* and R2 maps in the patient and healthy control. As shown in Figure 2, R2* values are markedly elevated in all basal ganglia regions of the patient, with the highest in the putamen (R2*= 125.3 ± 42.1 s⁻¹), corresponding to an average 3.14-fold increase over the control. In contrast, R2 values increase only moderately, with a mean factor of 1.57. Microstructural parameters from SDT are summarized in Table 1 and compared with R2 values from the SPACE sequence (R2SE) and QSM-based susceptibilities (X_QSM). Figure 3 shows the fitting curves for the putamen. In the patient, SDT-derived absolute susceptibilities (|X_SDT|) range from 0.176 ± 0.096 ppm in the pallidum to 0.226 ± 0.094 ppm in the putamen, consistently exceeding QSM values, which range from 0.01 ± 0.16 to 0.24 ± 0.33 ppm. In the control, this overestimation is even more pronounced. Notably, |X_SDT| values do not significantly differ between patient and control. The SDT model also revealed elevated volume fractions (η) of magnetic inclusions in the patient, exceeding 10% in all regions and peaking at 13.5 ± 8.0% in the putamen, compared to an average of 2.95% in the control. In nearly all regions, SDT yielded a lower mean squared error (MSE) than the mono-exponential model, except for the control’s caudate. Our findings suggest that the altered transverse relaxation observed in the basal ganglia of ACP patients arises primarily from the inhomogeneous distribution of magnetic susceptibility sources, rather than solely from elevated iron concentrations. The notable difference in R2*/R2 ratios between groups indicate that static dephasing is the dominant signal decay mechanism in ACP. This is supported by the post mortem observation of clustered ferritin-bound iron aggregates, generating localized susceptibility gradients [1, 2]. Volume fractions over 10% in regions like the putamen highlight clustered iron buildup in ACP. Similar vascular contributions in patients and controls make vascular changes unlikely to explain the pronounced contrast. These findings support ferritin aggregation as the main source of susceptibility alterations in ACP. This study demonstrates the utility of SDT-based modeling of iron related microstructural alterations in ACP. Beyond mapping the spatial distribution of ferritin-bound iron, this method allows quantification of microstructural biomarkers directly from the MR signal decay. Such advancements will allow new mechanistic insights into ACP pathogenesis.
Alexander STÜRZ (Innsbruck, Austria), Marlene PANZER, Heinz ZOLLER, Elke R. GIZEWSKI, Christoph BIRKL
15:30 - 17:00
#45945 - PG298 Radiomic Feature Robustness Affected by Magnetic Resonance Field Strength for Patients with Alzheimer’s Disease.
PG298 Radiomic Feature Robustness Affected by Magnetic Resonance Field Strength for Patients with Alzheimer’s Disease.
Alzheimer's disease (AD), an irreversible neurodegenerative disorder affecting over 55 million people worldwide[1], presents a critical public health challenge intensified by aging populations. Advances in neuroscience and radiomics now enable the systematic tracking of AD progression through conventional MR images[2]. Recent studies have increasingly focused on evaluating the impact of MRI field strengths on diagnostic performance, particularly modeled as classification tasks[2-4]. The radiomics features extracted from MR images, correlating with hippocampal atrophy and cortical thinning, may be considered valuable biomarkers reflecting pathological and functional changes.
However, a critical gap persists in understanding how individual MRI-derived features behave across field strengths. In brain imaging, MR images are acquired using different manufacturers, magnetic field strengths, and acquisition parameters. These variations can significantly affect further radiomics studies, especially different magnetic field strength. In this study, our objective was to assess the robustness of specific radiomic features across different MRI field strengths and evaluate their diagnostic value for AD detection.
This retrospective study analyzed 253 subjects from the ADNI database (https://adni.loni.usc.edu) with paired 1.5T and 3T MRI scans . Of these, 191 subjects were included because their scans were conducted within a 3 month period: 41 Alzheimer’s disease (AD) patients, 88 mild cognitive impairment (MCI) patients, and 62 cognitively normal controls (NC) (age 74.67 ± 7.21 years; 95 males/96 females). All scans utilized standardized T1-weighted sequences with harmonized parameters across scanners/sites under quality control at the Mayo ADIR lab.
Bilateral hippocampi were segmented based on HippoDeep method[5] for 1.5T and 3T scan respectively. Subsequently, radiomics features were extracted from the segmented hippocampi using Pyradiomics[6]. To evaluate the reproducibility and robustness of the extracted features across 1.5T and 3T scans, the nonlinear concordance correlation coefficient (NCCC) and the Wilcoxon signed-rank test (WSR) were used.
The acceptable stable features (NCCC > 0.6) were used for classification tasks to distinguish between NC and MCI, as well as between MCI and AD. Rather than using the features that passed the Wilcoxon signed-rank test, the ten most informative features were selected based on mutual information scores. A Random Forest classifier was trained on the training set (80%), and its performance was evaluated on the testing set (20%). To enhance robustness and reduce the risk of overfitting, five-fold cross-validation was performed within the training set during model development. Figure 1 shows representative hippocampal segmentation from NC, MCI, and AD subjects' MRI at both 1.5T and 3T field strengths. Among the 120 radiomic features, 27 features displayed acceptable concordance between 1.5T and 3T (NCCC > 0.6). Most of the features were shape-related, as shown in Figure 2. Figure 3 shows the classification performance based on area under curve (AUC) for distinguishing NC-vs-MCI and MCI-vs-AD groups at both 1.5T and 3T field strengths. In this work, several stable features were identified that were not significantly affected by field strength and were able to preserve hippocampal characteristics across different field strengths. Using stable features, four Random Forest classifiers were constructed to successfully classify subjects into NC, MCI, or AD. These models achieved above 80% area under curve (AUC) for MCI-vs-NC, i.e. strong discrimination. However, differentiating MCI from AD remains challenging, with 71% and 76% AUC, respectively for the 1.5T and 3T MRIs. Potential reasons for this are that i) MCI encompasses diverse range of microstructure changes, making it a complex condition to classify, and ii) subtle features captured by NCCC are insufficient to detect changes in MCI progression. Therefore, future research may need to incorporate more refined MCI stratification strategies and employ enhanced feature techniques for comparing feature robustness to improve the classification performance between MCI and AD. The common radiomics features across 1.5T and 3T demonstrate potential for distinguishing NC, MCI and AD. Additionally, those stable features truly possess generalizable diagnostic value for early Alzheimer's disease.
Jiqing HUANG (Liège, Belgium), Yi CHEN, Christophe PHILLIPS
15:30 - 17:00
#47792 - PG299 Mapping perfusion in thalamic nuclei of cognitively normal older adults using high-resolution ASL at 3 T.
PG299 Mapping perfusion in thalamic nuclei of cognitively normal older adults using high-resolution ASL at 3 T.
Aging has been known to show changes in cerebral perfusion (Liu et al. 2012) that has led to many large consortia acquiring ASL data to non-invasively map perfusion. However, these studies have acquired data at very low-spatial resolutions (≥3.5mm in-plane and 5-7mm slice thickness). Whilst this may be sufficient to map cortical perfusion and age- and disease-related perfusion changes in at the scale of whole brain areas, it is insufficient to resolve (Kashyap et al. 2024) smaller, deeper brain areas such as hippocampus, thalamic nuclei that are known to be impacted in disorders such as Alzheimer’s (Rodriguez et al. 2000) or Parkinson’s (Kimura et al. 2011). For this ongoing cross-sectional study, we optimised a high-resolution ASL protocol (2.5 mm iso) to acquire CBF maps in clinically-feasible times (< 6 min) at 3 T, and demonstrate its utility to capture perfusion-changes in thalamic nuclei in a cohort of cognitively normal older adults.
2.5 mm isotropic 3D-GRASE pCASL (TR/TE=3.93s/20ms, GRAPPA 3, PF=6/8, Label=1.8s,PLD=1.8s,TF=12,Seg=4,EPI Factor=27,BW=2480Hz/Px, TA=5:42 min) was acquired in 35 healthy older adults (24F,11M) aged 56.0 to 84.5 years (mean ± SD = 71.9 ± 8.1 years) on a Siemens Prisma 3T (Siemens Healthineers, Erlangen). Data were processed using oxasl (Chappell et al. 2023) and THOMAS (Su et al. 2019) was used to obtain delineations of thalamus and its nuclei. ANTs (Tustison et al. 2021) was used for all registrations and cohort template building. Group average THOMAS parcellation (Fig 1a) and corresponding volumes (Fig 1b) are consistent with Su et al. 2019. Perfusion-weighted image and CBF maps (Fig 1c) show observable differences in the thalamus (white outline) and the mean CBF (Fig 1d) differed markedly across thalamic subregions. The habenula exhibited the lowest perfusion (~35 ml/100 g/min), whereas the anteroventral and mediodorsal nuclei showed the highest (~73 ml/100 g/min). Most nuclei fell within one standard deviation of the global thalamic mean (~56 ± 23 ml/100 g/min) and show negligible lateralization. There was a non‐significant mild positive correlation between nuclei volume and mean CBF (r(10) = 0.50, p = 0.098, R² = 0.25), indicative of a trend toward higher perfusion in larger nuclei. A non-significant inverse relationship between age and perfusion (slope ≈ –0.15 ml/100 g/min per year) can be observed (Fig 1e) consistent with expectations in healthy aging. Region‐specific trends are similar between sexes, with females having consistently higher CBF by ~5–15 ml/100 g/min (Fig 1f). We demonstrate the feasibility of a 2.5 mm isotropic 3D-GRASE pCASL protocol at 3 T to resolve perfusion differences across individual thalamic nuclei in a clinically acceptable scan time (< 6 min) in 35 cognitively normal older adults. We show an approximately two‐fold range in baseline CBF across nuclei (Hb ~ 35 ml/100 g/min to AV and MD ~ 73 ml/100 g/min). The observed perfusion differences indicate differing cytoarchitecture and metabolic demands of thalamic subregions. These results, part of an ongoing clinical study, establishes a normative baseline to compare subregional thalamic dysfunction in early diagnosis and monitoring of disease progression in Alzheimer’s and Parkinson’s disease. We show high-resolution ASL at 3T overcomes the limitations of conventional ASL to non-invasively map perfusion and resolve CBF changes in small, deep grey-matter structures such as thalamic nuclei, and demonstrate the capability to acquire them in clinically-feasible times.
Sriranga KASHYAP (Toronto, Canada), Kevin SOLAR, Nicolas DEOM, Josef PFEUFFER, Mary Pat MCANDREWS, Kamil ULUDAG
15:30 - 17:00
#47307 - PG300 Reliability of MRI-derived multispectral neuroimaging measurements after 3T scanner relocation.
PG300 Reliability of MRI-derived multispectral neuroimaging measurements after 3T scanner relocation.
Ensuring the reliability of metrics derived from Magnetic Resonance Imaging (MRI) data is essential for both clinical and research applications. Scanner relocation to a new facility poses a potential challenge to the stability of MRI-derived data [1]. This study assessed the reproducibility of multispectral neuroimaging measures including diffusion, resting-state networks (rs-fMRI), task-based fMRI [2], and morphometry acquired before and after relocation of a 3T MRI scanner, in the absence of any software or hardware upgrades.
Six healthy volunteers completed 4 MRI sessions in a test-retest design before and also after scanner relocation (3T Siemens Prisma, 64 channel head-neck receive coil). Each session included: T1w Multi-Echo MPRAGE (meMPRAGE), Multishell DWI, UMN CMRR multi-echo EPI for rs-fMRI, single-echo EPI for task-based fMRI. Assessed metrics were: segmentation-based cortical and subcortical gray matter (GM) volume [3]; Mean-Signal Kurtosis (MSK; MS Diffusion Kurtosis Imaging, MS-DKI [4]) in GM and white matter [5] regions of interest (ROIs); strength and topology of the rs-fMRI Default Mode Network (DMN); Euclidean distance between functional peaks in ROIs based on contrasts mapping auditory areas and ventral/dorsal visual areas. Data were compared across the four sessions in terms of: absolute values, relative change, and reliability in all the considered metrics. Brain morphometry reproducibility (Figure 1): Two-way ANOVA on T1w structural data showed no significant results both for main effects (Session, Site) and their interaction, in any of the considered ROI (pFDR=NS). Relative changes remained well below the 5% threshold for within- and between-sites test-retest. One-way ANOVA on relative variations was non-significant. ICC(3,1) revealed excellent reliability (average ICC=0.98; CI(95%)=0.94-0.99).
Brain microstructure reproducibility (Figure 2). Relative MSK changes were below 3% across Sites and Sessions, with excellent reliability (ICC(3,1)=0.99; CI(95%)= 0.98-0.99). No site effect was observed in any ROI (all padj>0.3).
Intrinsic functional connectivity reproducibility (Figure 3). Resting-state connectivity analysis on average Z-score extracted from DMN revealed no significant differences for both main effects and their interaction in a two-way ANOVA. A further one-way ANOVA on relative changes was non-significant. Test-Retest measurements assessed with ICC(3,1) showed limited variability with an estimate of 0.99 (CI(95%)= 0.9-1), in good agreement with literature [6]. Spatial distribution of DMN was also tested across Sites and Sessions: non-significant differences were reported with TFCE-corrected non-parametric permutation testing.
Task-based fMRI reproducibility (Figure 4). The Euclidean distance between two selected peaks in the ROI analysis—performed separately for the auditory–visual and visual–auditory stimulus contrasts—revealed no significant main effects (Site, Sessions) or interaction (two-way ANOVA, p=NS). Moreover, the two one-way ANOVAs (one for each contrast) on the relative change of the Euclidean distance were not significant. On average, the relative variation of the Euclidean distance between peaks remained below the 5% threshold for both within- and between-sites comparisons. Finally, the two ICC(3,1) performed on the Euclidean distance in both contrasts indicated a good reliability, with estimates of 0.60, (CI(95%)=0.3-0.8) and 0.70 (CI(95%)=0.5-0.9), respectively. By systematically comparing pre- and post-relocation data within the same healthy volunteers, we aimed to determine if scanner relocation introduces biases in the considered metrics. Our data show high consistency across all examined measurements. Despite technical changes, such as hardware calibration, and differences in environmental factors like room characteristics and ambient conditions, we did not observe any meaningful variation in the results. Test-retest reproducibility within- and across-site locations were not significantly different. Scanner relocation had minimal impact on our neuroimaging measurements: structural measures varied <5% (ICC = 0.98), diffusion MSK <3% (ICC = 0.99), task-based fMRI activation peak distances <5% (ICC = 0.60-0.70). A high overall reproducibility (ICC = 0.99) was found even for resting-state DMN topology. These results confirm the robustness of MRI neuroimaging measures post-relocation.
Alberto FINORA (Rovereto, Italy), Stefano TAMBALO, Paula Andrea MALDONADO MOSCOSO, Sebastian HÜBNER, Lara Maria VIOLA, Jorge JOVICICH
15:30 - 17:00
#47855 - PG301 The EEG-fMRI temporal correlations of motor brain activity vary over time and between task and rest conditions.
PG301 The EEG-fMRI temporal correlations of motor brain activity vary over time and between task and rest conditions.
Simultaneous electroencephalography (EEG) - fMRI acquisitions are used to investigate brain activity due to the highly complementary characteristics of the two neuroimaging modalities [1]. EEG-fMRI studies of the sensorimotor network (SMN) are of particular interest for applications in the neurorehabilitation of motor function, such as neurofeedback (NF) during motor imagery (MI) [2]. However, studies comparing EEG fluctuations with concurrent SMN fMRI activity have reported inconsistent findings, typically showing only very low-amplitude temporal correlations [3]. We hypothesize that this may partly stem from fluctuations in these correlations over seconds to minutes and also across different conditions. Here, we investigate the within-subject dynamics of temporal correlations between SMN fMRI and concurrent EEG spectral power across both task and rest conditions.
Simultaneous EEG-fMRI data were recorded from 15 healthy subjects in 2 sessions ~2 weeks apart. In each session, subjects participated in two ~17min MI tasks (Graz and NeuRow) and a 10min rest scan with eyes open (Rest). fMRI data were acquired on a 3T Siemens system using 2D EPI (TR/TE=1260/30ms, 60 axial slices, 2.2mm iso). EEG data were collected using a 32-channel MR-compatible BrainAmp system, with 31 EEG channels and 1 ECG channel, synchronized with the fMRI scanner. Regarding MI tasks, after standard preprocessing, a GLM analysis was conducted to identify the SMN and extract its average BOLD signal. A BOLD time series representative of the spontaneous trial-by-trial fluctuations (TBT) was also obtained by regressing out the task-related effects. For Rest, after standard preprocessing and denoising, a group-level ICA was applied to identify the SMN, which was back-projected to individual subjects to obtain subject-specific BOLD time series. After artifact removal and standard preprocessing, EEG data underwent time-frequency decomposition using Morlet wavelets (3s temporal, 1Hz frequency resolution) to obtain the relative power across frequencies and time points for each channel. These power time series were downsampled to match the temporal resolution of the BOLD signal using an anti-aliasing low-pass filter. For each condition (Graz-TE, NeuRow-TE, Graz-TBT, NeuRow-TBT, Rest), subject, and session, sliding windows of 36 TRs with ~90% overlap were applied to EEG and fMRI data. For each sliding window, Pearson correlation was computed between the SMN fMRI time series and EEG power across 40 frequency bins (1-40Hz) and 31 channels, considering 9 time lags (0-8 TRs~10s). The time lag yielding the strongest correlation was selected for each condition, subject, session, channel, frequency bin, and sliding window. K-means clustering was used to identify recurrent correlation patterns, exploring cluster numbers from k=2 to k=8, with further analysis revealing 4 distinct states. Fig.1 displays the centroids of the 4 states (A, B, C, and D) found in the EEG-fMRI correlation dynamics for each condition. Fig.2 shows time-dependent EEG-fMRI correlations for Graz-TBT, averaged across channels and concatenated across subjects and sessions for each connectivity state. Fig.3 presents the Euclidean distances between EEG-fMRI correlation states for different conditions. States A and B demonstrated consistent patterns across all conditions, as reflected by their lower Euclidean distances between conditions. In contrast, states C and D exhibited greater variability, with state C showing the largest distances in most cases. Fig.4 illustrates the temporal dynamics of the states’ parameters, including dwell time and fractional occupancy. A two-way repeated measures ANOVA revealed a significant main effect of state for both dwell time (F=2.97, p=0.04) and fractional occupancy (F=4.1, p=0.01). Additionally, a significant interaction between state and condition was observed for both measures (dwell time: F=7.3, p<0.001; fractional occupancy: F=3.4, p<0.001). Post-hoc analyses, adjusted with Bonferroni correction for a family of 40, were conducted to explore the significant (p<0.05) 2-way interaction identified between state and condition. Differences in dwell time between conditions were found for states A, B and C. No significant effects were found for fractional occupancy in the post-hoc comparisons. While states A and B displayed symmetric correlation patterns across a broad frequency band above 10 Hz, states C and D showed symmetric correlation patterns centered around the alpha band near 10 Hz. The correlation patterns remained consistent across time windows for each connectivity state, suggesting stable EEG-fMRI coupling within each state. Dwell time results show that the stability of each state varies depending on the conditions. Our results reveal high variability in EEG-fMRI correlations in the SMN over time and across different task and rest conditions, offering a potential explanation for the consistently low-amplitude correlations reported in the literature.
Joana MENDES (Lisbon, Portugal), Marta XAVIER, Neil MEHTA, Athanasios VOURVOPOULOS, Patrícia FIGUEIREDO
15:30 - 17:00
#47447 - PG302 Threshold PCA denoising outperforms MP-PCA in Correlation Tensor Imaging data of human brain microstructure at 3T.
PG302 Threshold PCA denoising outperforms MP-PCA in Correlation Tensor Imaging data of human brain microstructure at 3T.
Correlation Tensor Imaging (CTI) is an advanced diffusion MRI method based on Double-Diffusion-Encoding (DDE) that separates diffusional kurtosis into isotropic (Kiso), anisotropic (Kaniso), and microscopic (μK) components, offering greater specificity for brain microstructure characterization [1]. While CTI has shown strong potential in preclinical studies [2], and was recently demonstrated in humans [3], current protocols require long acquisitions (~50 min), limiting clinical feasibility. Accelerating CTI will drastically reduce acquisition time but also exacerbate signal-to-noise ratio (SNR) losses, making effective denoising crucial. PCA-based methods such as Marčenko-Pastur PCA (MP-PCA) [4] and Threshold PCA (TPCA) [5] have shown promise for denoising diffusion MRI data without compromising microstructural information. While MP-PCA denoising classifies principal components based on the Marčenko-Pastur distribution, assuming spatially uncorrelated noise, TPCA incorporates a prior estimate of the noise variance to establish a threshold for noise component removal, enhancing robustness in scenarios with spatially correlated noise. This study evaluates, for the first time, the effects of MP-PCA and TPCA denoising on CTI-derived metrics in healthy human brains.
Population & MRI Acquisition: We used CTI data acquired from 8 healthy young volunteers, as described in a previous 3T study [3].
Preprocessing and CTI: Three CTI preprocessing pipelines (P1–P3) were implemented, differing only in their PCA denoising approach: P1 – no denoising; P2 – MP-PCA (MRtrix3) [4]; P3 – TPCA (Dipy) [5]. After denoising, all pipelines included the same subsequent corrections in the following order: Gibbs ringing (MRtrix3), geometric distortions and eddy currents (FSL), signal drift [6], and bias field (MRtrix3). CTI metrics were fitted and estimated within different brain regions of interest (ROIs).
Statistical Analysis: To assess the impact of denoising across different processing pipelines, we compared three key metrics within ROIs: 1) the mean CTI estimates, 2) the within-ROI variability of CTI values, and 3) the percentage of voxels with biologically implausible (i.e., negative) CTI fits. Statistical analysis was conducted using within-subject repeated measures. A Friedman test was first applied to identify overall effects of the pipeline. When significant, this was followed by right-tailed Wilcoxon signed-rank tests for pairwise pipeline comparisons. P-values were adjusted for multiple comparisons using the False Discovery Rate (FDR) method. Figure 1 shows sample subject results from the three pipelines on CTI metrics. Denoising improvements are most visible on μK. At the group level, there were no significant differences across pipelines on CTI metrics averaged in the ROIs. When looking at within-ROI variability and percentage of negative CTI fits, we observed significant improvements with denoising, in particular with TPCA (Table 1). Figure 2 shows how voxel-wise variability of CTI metrics is systematically and significantly reduced within the various ROIs considered when moving from P1 to P2 to P3, especially for μK (p < 0.01). In addition, we found similar effects of denoising improvements when looking at the percent of negative CTI model fits (biologically impossible values) within the various ROIs, especially for μK (Figure 3). Our main findings are twofold. First, PCA-based denoising significantly reduces within-ROI variability of CTI metrics and decreases the number of voxels yielding biologically implausible model fits. Second, these denoising effects are markedly stronger when using TPCA compared to MPPCA. Both effects were consistently observed across gray and white matter regions in healthy young brains. CSF showed the strongest denoising improvements in terms of reduction of negative fits, maybe related to the fact that Rician denoising was not included. These preliminary results highlight the value of incorporating PCA denoising strategies into the human CTI protocol at 3T to enhance data quality and reliability, in particular TPCA. While these findings are promising, further work is needed to further characterize CTI denoising optimization for clinical and research applications. Future studies will investigate the additional impact of Rician bias correction within the CTI framework on a larger sample, as well as assess how these denoising approaches influence test-retest variability, an important marker of reproducibility.
Manu RAGHAVAN (Rovereto, Italy), Lisa NOVELLO, Rafael Neto HENRIQUES, Noam SHEMESH, Andrada IANUŞ, Thorsten FEIWEIER, Domenico ZACÀ, Jorge JOVICICH
15:30 - 17:00
#47570 - PG303 Spatial Pointwise Orientation Tracking (SPOT): Resolving the spatial layout of fibre ODFs for super-resolution streamlining.
PG303 Spatial Pointwise Orientation Tracking (SPOT): Resolving the spatial layout of fibre ODFs for super-resolution streamlining.
A fundamental and hitherto unsolved challenge in diffusion MRI tractography is to uncover the spatial distribution of orientations within an imaging voxel from the voxelwise fibre orientation distributions (FODs). Streamlining in tractography attempts to solve this problem through a combination of interpolation and heuristics [1], such as penalising sharp turns in the streamlining. Although this approach has been successful over the years in mapping large coherent bundles, tractography notoriously still suffers from "bottleneck" issues where the FOD can ambiguously represent different underlying orientation configurations [2]. The problem is that once we've built an FOD, it is too late to recover the underlying pointwise orientations.
Here we propose a potential paradigm shift: rather than estimating pointwise orientations through streamlining after fitting FODs, we propose to model the underlying pointwise orientations directly in a forward model of the data that bypasses the need for fitting FODs (Fig 1A). Pooling the data across neighbourhoods of voxels makes this model tractable (invertible). We present a preliminary version of the model in simulated and real data, and suggest how this type of approach can be used for joint modelling of dMRI and polarised light imaging (PLI) data from the same tissue, where the PLI further constrains the model.
At the core of the forward model are simple multi-layer perceptrons (MLPs, which in these experiments have 4 layers, 256 neurons per layer, tanh activations, linear output). The MLPs take as input the (x,y,z) coordinates of any point in space (not necessarily at the centre of a voxel), and output either a 3D vector (for modelling the orientation vector field) or a scalar (for modelling e.g. the diffusion coefficient). To enable modelling of both high and low frequencies in the spatial domain, we insert a Fourier Feature embedding layer between the input and the MLPs [3]. The outputs of the MLPs (for the vector and scalar fields) are then used within forward models of the data (Fig 1B). For example, to predict diffusion data, sample positions from within a voxel are fed through the MLPs to generate orientations and scalars, then fed through the diffusion model equation for a stick given a set of bvals/bvecs, and then averaged over the voxel. The predicted signal is then compared to the measured diffusion data to calculate a mean squared error loss. This is summed over a neighbourhood of voxels to calculate the overall loss used for training the MLPs. Other data, such as polarised light imaging (PLI), can be similarly predicted from the orientation vector field, enabling the joint modelling of multiple modalities (optional extra). We call this method SPOT (Spatial Pointwise Orientation Tracking).
SPOT was evaluated using simulated diffusion data from an orientation vector field (here 2D) which we aimed to recover. For comparison with interpolation, the data was resampled onto a 100x100 grid using splines of order 3 (with Scipy's map_coordinates), and a tensor was fitted in each pixel to obtain a principal orientation per pixel.
Next, we assessed the impact of jointly fitting a single dMRI voxel alongside one or two slides of high-resolution PLI intersecting the voxel. Finally, we show some early results fitting SPOT to in vivo dMRI data (60 directions, 2mm isotropic, b=1k). Preliminary results are shown in figures 2-4. In 2D simulations, we compare SPOT to spline interpolation (Fig 2) for different grid sizes (1x1, 2x2 and 4x4) to demonstrate the effect of including neighbourhood information. SPOT outperforms interpolation in terms of the resolved pointwise orientations and the FODs. When jointly fitting to MRI and PLI (Fig 3), SPOT manages to recover the underlying pointwise orientations from a single voxel (no neighbourhood information) even with a small amount of PLI as additional constraint . Results comparing SPOT to constrained spherical deconvolution (CSD) in in vivo data (Fig 4) show how FODs with crossing fibres resolve into streamlines bending onto the cortex in SPOT. SPOT is still very much under development. Future work includes: investigation of the effect of spatial resolution, b-values, inclusion of multiple modalities, and size of the spatial neighbourhood. Current implementation is limited to a small neighbourhood, and further work is needed to exploit the pointwise vector field for mapping long-range connectivity. SPOT is a super-resolution, multi-modal technique that attempts to recover the pointwise orientation (and other scalar) field(s) directly from voxelwise dMRI data.
Saad JBABDI, Amy HOWARD (London, United Kingdom)
15:30 - 17:00
#47854 - PG304 Variability of individual and group-level functional brain network hubs as a function of parcellation schemes and network density.
PG304 Variability of individual and group-level functional brain network hubs as a function of parcellation schemes and network density.
Graph theoretical analysis of brain networks is a powerful method for quantifying topological features that may help differentiate between healthy and disease-affected brains. During analysis of networks based on resting-state functional MRI (rs-fMRI), nodes are defined as parcellation-derived regions of interest (ROIs) and edge weights as temporal correlation coefficients. [1] Analyzing neuroimaging-derived networks is hindered by the multiplicity problem, the unclear nature of data distribution and having insufficient prior knowledge on the location of effects of interest or the core connectome structure. [2,3]
We investigated rs-fMRI derived functional brain networks of controls and patients diagnosed with epilepsy to better understand the core structure and topology of these networks and also the effects of thresholding on graph theory metrics. We aimed to identify regions that can be considered hubs in healthy controls and investigate how hub-identification depends on network thresholding and the fineness of the ROI-resolution.
Rs-fMRI of 28 healthy controls and 34 patients diagnosed with epilepsy were acquired on the 3T Philips Ingenia scanner of Semmelweis University Medical Imaging Centre (TR=1s, 2.5mm isotropic resolution, 260 volumes). Corresponding structural images were segmented in FreeSurfer7.4.1 with 5 ROI sets: 84 ROIs defined following the Desikan-Killiany atlas (DK); a finer parcellation of 360 cortical ROIs (Glasser); one with 14 subcortical DK-ROIs (Glasser-DK); 168 ROIs of the AAL3 parcellation; and 70 subcortical of these added to Glasser's (Glasser-AAL). [4] After preprocessing with CONN22.a, individual ROI-to-ROI connectivity matrices (RRCs) were calculated for all 5 ROI-sets, then thresholded by density. [5] We calculated graph metrics for each network using Brain Connectivity Toolbox (BCT). Centrality metrics and clustering of nodes were weighed by ROI-sizes. Nodes with the highest 15% of a given metric were defined as subject-level hubs or highly clustered regions. Group-level hubs were defined if the overall median metric value was in the top 15% among all ROIs. Ratios of differences between individual and group-level hubs were compared across thresholds between controls and patients. Overlap of group-level and individual hubs was assessed through metrics integrated over density ranges. Nonparametric Mann-Whitney tests (α=0.05) were used for between-group comparisons. Individual hubs showed large variance for both groups, with 58-75% difference from group-level hubs for all centrality metrics and thresholds. The differences decreased with higher network density for degrees, betweenness and eigenvector centralities in coarser parcellations (DK, AAL). Differences decreased in the control group, but increased in the patient group for strengths, and degrees in finer parcellations (Glasser-based – Fig. 1). Overall, we found statistically significant differences between the strength-based hubs of the patient and control groups at high network densities (>26%) and fine parcellations, see Fig. 2. Group differences in terms of highly clustered regions substantially decreased with rising network densities, yet significant differences were found between the groups at multiple densities, mostly in finer parcellations. Increasing network density had the most dramatic effect on results with the AAL-set, both for centrality metrics (exception for strength) and clustering. This may be due to large size differences between ROIs in this set, which distorted metric scaling.
Based on small-worldness, we identified three threshold ranges: at 1-5% density, networks were sparse, between 6-18% they changed rapidly and around 19% they stabilized. We investigated overlaps between subject and group-level hubs by integrating metrics on the three threshold-ranges. ROIs corresponding to parts of the default-mode network (DMN) were denoted as hubs at all density ranges and ROI-sets. Other consistently identified hubs included inferior parietal areas, superior temporal gyri, frontal and parietal opercular regions. Parts of the middle frontal gyri were also frequently identified as hubs. Medial areas of the DMN, unimodal visual, primary auditory and opercular areas were identified as highly clustered regions. Bilateral parahippocampal regions showed high clustering in Glasser-based parcellations at all densities but only appeared at higher densities in the DK-parcellation. Our results are compatible with pertinent literature regarding group-level hubs [6], as identified areas that overlap with the DMN and association regions. Interestingly, auditory areas are identified both as hubs and highly clustered regions, a possible effect of scanner noise. Strength-based hubs in networks derived from finer parcellations and higher than 26% density were found useful to identify between-group differences. Confirmation of these results on other patient groups with possible inclusion of other weighted metrics seems beneficial.
Noémi GYÜRE (Budapest, Hungary), Gyula GYEBNÁR, Lajos Rudolf KOZÁK
15:30 - 17:00
#47640 - PG305 CACTUS 2.0: Towards a Hierarchical Framework for Generalized Neural Tissue Modeling.
PG305 CACTUS 2.0: Towards a Hierarchical Framework for Generalized Neural Tissue Modeling.
Monte Carlo simulations are essential for analyzing and validating microstructural models in Diffusion-Weighted Magnetic Resonance Imaging (DW-MRI), especially when analytical solutions are not available. These simulations require accurate geometric substrates to represent the underlying tissue microstructure [1,2,11] realistically. Over the years, the research community has proposed several substrate generation methods for white matter (e.g., CACTUS, Medusa) and gray matter (e.g., CONCEG), offering varying levels of anatomical detail and biological realism [3–5,9]. In this work, we present CACTUS 2.0, an enhancement of the original CACTUS framework, which was designed for ultra-dense packing of simplified white matter substrates. CACTUS 2.0 extends these capabilities to support hierarchical cellular structures, enabling the modeling of complex, branching obstacles such as neurons. This development retains the high-density packing strengths of the original system, achieving volume fractions of up to 70% in gray matter–like configurations.
This tailored enhancement of the original framework introduces three key features: (1) cellular packing optimization using density-preserving deformation algorithms, (2) hierarchical branching systems with controllable morphology, and (3) integration of pre-segmented neuronal structures.
To achieve high cellular densities (up to 95%), we adapted metaball-based techniques with dynamic growth factors that enable efficient packing. For modeling hierarchical structures, we implemented a recursive branching algorithm that supports user-defined control over morphological parameters such as branching angle, segment length, diameter, and distribution. This allows the generation of diverse structural patterns, including pyramidal, targeted, and omnidirectional growth. Lastly, the CACTUS optimization framework was extended to account for complex hierarchical geometries while preserving their morphology during collision resolution and overlap removal. CACTUS 2.0 successfully generated dense and morphologically realistic cellular substrates. As shown in Figure 1, our growth-based optimization outperforms traditional sphere packing methods, which plateau around 55% density, by reaching up to 95% packing density, producing biologically plausible, non-spherical deformations that align with electron microscopy observations [8].
Figure 2 highlights the versatility of our framework in handling diverse neuronal morphologies. It includes synthetic pyramidal neurons representative of neocortical layers 2–3 (a,b), targeted structures with directional bias (c,d), and isotropic omnidirectional growth (e,f), as well as segmented real neurons (g) from neocortical tissue [6,7].
Figure 3 demonstrates the ability to preserve hierarchical morphology around large obstacles, emulating arrangements such as astrocytic wrapping around somas.
Figure 4 shows a full-scale substrate integrating 350 neurons (based on real templates), within a 200 μm × 200 μm × 100 μm volume. After optimization, all overlaps are resolved, and the resulting packing density reaches 70% in the central part of the voxel, a benchmark previously out of reach for neuron-level simulators. CACTUS 2.0 significantly expands the modeling scope of substrate generation tools by incorporating hierarchical, biologically-inspired structures such as neurons and glia. By supporting synthetic and segmented cellular morphologies, the framework enables the construction of ultra-dense and structurally diverse substrates suitable for simulating gray matter microenvironments.
The primary challenge now shifts from achieving dense packing to reproducing biologically realistic spatial arrangements across brain regions. Although CACTUS 2.0 can efficiently integrate microscopy-derived structures, capturing region-specific patterns—including accurate soma placement, branching orientation, and local cellular diversity—requires further anatomical guidance. Solving these new challenges will unlock a new potential for using these substrates in geometric modeling and studies that demand functional fidelity, such as connectomics and DW-MRI signal interpretation. CACTUS 2.0 represents a substantial advance in computational modeling of brain tissue microstructure. Its capacity to achieve packing densities above 70% while preserving complex, hierarchical morphologies makes it a powerful tool for simulating tissue architectures relevant to diffusion MRI and microstructure-informed analysis. CACTUS 2.0 lays the groundwork for more realistic signal simulations and validation studies by enabling the generation of high-fidelity, voxel-scale substrates.
Juan Luis VILLARREAL HARO (Lausanne, Switzerland, Switzerland), Jean-Philippe THIRAN, Jonathan RAFAEL-PATINO
15:30 - 17:00
#46644 - PG306 White Matter Alterations in MCI Using Free-Water Corrected Tractography: A Comparative Study with Standard DTI.
PG306 White Matter Alterations in MCI Using Free-Water Corrected Tractography: A Comparative Study with Standard DTI.
Mild cognitive impairment (MCI) is a transitional stage between normal aging and dementia, particularly Alzheimer’s disease (AD) [1], marked by cognitive decline, especially in memory, without major disruption to daily life [2].
Diffusion MRI (dMRI) assesses white matter integrity [3], and previous studies using tractography have revealed reduced connectivity in MCI, particularly in memory- and attention-related pathways [4]. However, conventional diffusion tensor imaging (DTI) is limited in areas with free-water (fw), skewing metrics like fractional anisotropy (FA) and mean diffusivity (MD) [5]. Fw correction of multi-shell dMRI improves accuracy by separating tissue and fw signals [6]. However, few studies have examined the impact of fw contributions on tractography.
This study compared standard vs. fw-corrected dMRI (tractography and DTI metrics) using Alzheimer's Disease Neuroimaging Initiative (ADNI) data [7] from healthy controls (HCs) and MCI patients. We also assessed how diffusion directions affect tractography and explored structural connectivity differences, linking them to cognitive performance. By combining fw correction and multi-shell dMRI, this work provides deeper insights into white matter changes in MCI.
The study included 153 participants (105 HCs and 48 with MCI). Cognitive function was evaluated using the Mini-Mental State Examination (MMSE) [8], and apolipoprotein E (ApoE) status was assessed as a genetic risk factor for AD [9]. Multi-shell dMRI data were collected on a 3T Siemens scanner, with 127 diffusion directions at b-values of 500, 1000, and 2000 s/mm². Preprocessing steps included restructuring the dMRI data into subsets of 70, 90, and 110 directions to generate synthetic datasets, applying noise reduction, and performing free-water-corrected tractography analysis using a multi-tissue constrained spherical deconvolution approach [10]. Figure 1 summarizes the demographic, cognitive, motion, and APOE characteristics of the groups.
Synthetic dMRI datasets with varying gradient directions and noise levels were created to examine the effects of angular resolution and free-water correction on tractography accuracy. Structural connectivity differences between HC and MCI groups were analyzed using both standard and free-water-corrected tractography. Additionally, a region-of-interest (ROI) analysis was conducted on diffusion metrics for connections showing significant group differences. Statistical comparisons included paired t-tests (standard vs. corrected metrics across different directions) and independent t-tests (group differences in connectivity and ROI metrics), with sex, age, and motion as covariates. Effect sizes were computed using Cohen’s d (Figure 2). The fw model yielded significantly longer fiber lengths (p < 0.001) and lower false positive rates (p < 0.01), whereas the standard model produced shorter fibers and higher false negatives (p < 0.001). Overlap fraction decreased in some cases, particularly for the fw model at 70/90 directions, while orientation error remained similar across models.
Figure 3(a) displays standard tractography-derived connectivity differences between groups. The accompanying table reveals HCs showed stronger connectivity overall, except for the right path between Heschl’s gyrus and the right temporal pole, where MCI showed increased connectivity. Figure 3(b) presents the results using fw-corrected tractography, demonstrating uniformly higher connectivity in HCs, especially involving the left middle frontal gyrus (MFG.L).
Figure 4 compares standard DTI (a) and fw-corrected DTI (b) metrics between groups. No significant differences emerged with standard DTI, but fw-correction identified a higher fw-index in MCI for the right inferior frontal gyrus (IFGoperc.R) to right insula (INS.R) pathway (t = 3.155, p = 0.002), underscoring its enhanced sensitivity to microstructural changes. Both standard and fw-corrected tractography revealed reduced structural connectivity in MCI compared HCs. Notably, fw-corrected tractography demonstrated stronger statistical effects than standard methods, suggesting improved sensitivity to group differences.
While standard diffusion metrics failed to detect significant changes, the fw-corrected model identified a higher fw-index in MCI for the right inferior frontal gyrus (IFGoperc.R) to right insula (INS.R) pathway. This finding supports the hypothesis that fw correction enhances detection of early microstructural changes in white matter, which may precede overt cognitive decline. Fw-corrected tractography provides a more precise tool for assessing white matter degeneration in MCI compared to conventional DTI. The elevated fw-index in the IFGoperc.R–INS.R pathway—a region linked to cognitive control—underscores the potential of fw correction to uncover subtle neurodegenerative changes before they manifest clinically.
Maurizio BERGAMINO (Phoenix, USA), Marwan SABBAGH, Ashley M. STOKES
15:30 - 17:00
#47324 - PG307 Fast and robust SENSE reconstruction for spiral imaging.
PG307 Fast and robust SENSE reconstruction for spiral imaging.
Spiral trajectories provide a highly efficient strategy for spatial encoding of the object in MRI (see [1], [2]). However, in practice, image reconstruction of spiral scan data is challenging. Higher-order SENSE reconstruction [3] incorporates information about the spatial sensitivity profile of receiver coils, the static off-resonance field (B0) and higher-order gradient field perturbations. It is a generic approach as it renders image reconstruction as a linear inverse problem. However, generating accurate sensitivity maps and especially B0 maps difficult. We present two algorithms to address both issues. Although this type of SENSE reconstruction is computationally intensive, we demonstrate that fast image reconstruction is feasible using an optimized implementation running on modern GPUs.
Single subject data measured using the 16-channel Skope NeuroCam 3T (NeuroCam 3T, Skope Magnetic Resonance Technologies AG, Zurich, Switzerland) on a Philips Achieva scanner (31mT/m gradient strength, 200T/m/s slew rate, Philips Healthcare, Best, The Netherlands) has been used for all analysis/results presented in the following.
Both sensitivity and B0 maps are derived from images acquired during the same prescan. The corresponding sequence parameters used are provided in Figures 1 and 2. Following the reconstruction of single-coil images, the first temporal eigenvariate is removed to generate the initial sensitivity maps. This removed eigenvariate, which represents a coil-combined image, is also used to compute the trusted and recon mask. The trusted mask includes voxels with sufficient SNR to reliably estimate sensitivity maps, while the recon mask includes all voxels which might contain the object. The smoothing and extrapolation algorithm is then applied, enforcing consistency with the initial sensitivity maps within the trusted mask and penalizing the second derivative in all directions within the recon mask (see Figure 1).
The sensitivity maps are used to compute complex-valued, coil-combined images from the prescan data. An initial B0 map is then generated by performing a linear fit to the phase evolution across different echo times. The smoothing and extrapolation algorithm enforces consistency with the initial B0 map and penalizes the first derivative, in all directions (see Figure 2). The strength of this regularization, or penalty, is made locally proportional to the standard error of the linear fit. This formulation makes the algorithm preserve edges at location where the initial B0 map is accurate. Hence it behaves similar to total variation denoising (see [4]). However, it only requires the solution of a linear system of equations.
Within higher-order SENSE reconstruction a large linear system of equations has to be solved using the iterative CG-method [5]. A GPU-optimized implementation is used the reconstruct the 2D spiral scan data (see Figure 3). All allocated variables simultaneously fit into the memory of a commercially available GPU. No recomputation of the encoding matrix during each CG-iteration, as suggested in [3], is necessary. Most operations involving vectors and matrices can be highly parallelized on a GPU which results in a considerable speed-up. The smoothing and extrapolation of the sensitivity maps and the B0 map is depicted in Figures 1 and 2. Both smoothed and extrapolated maps look realistic and appear to have an increased SNR.
We consider the average runtimes of the most demanding operations indicated in Figure 3 as exceptionally fast.
Multiple reconstructed images are shown in Figure 4. Image quality is comparable to what is presented in [1]. The smoothing and extrapolation algorithms are capable of either sensitivity and B0 map enhancement, enabling high-performance SENSE reconstruction. Since SENSE reconstruction relies on an explicit signal model of the measured coil data, we suspect the computed maps to be physically meaningful and realistic.
Although higher-order SENSE reconstruction is computationally intensive, the GPU-optimized implementation enables fast image computation, effectively alleviating a key limitation of this method. The cost of the used GPU is negligible compared to the cost of an MRI scanner or an NMR field camera. However, for 3D spiral imaging the available memory might not suffice yet.
High-quality images were reconstructed even in challenging scenarios. The long acquisition time of 71ms makes the R=2 spiral scan particularly sensitive to local off-resonance effects, while the high undersampling factor of R=6 could introduce aliasing. Nevertheless, these typical artifacts are hardly visible in the reconstructed images. Using accurate sensitivity and B0 maps high quality single shot spiral images can be reconstructed. SENSE reconstruction can be accelerated using an GPU-optimized implementation. Our work makes spiral imaging using SENSE reconstruction practical.
Samuel BIANCHI (Zürich, Switzerland), Klaas P. PRUESSMANN
15:30 - 17:00
#47862 - PG308 Self-supervised UNet denoising algorithm for Rician bias correction of DWI data.
PG308 Self-supervised UNet denoising algorithm for Rician bias correction of DWI data.
Diffusion weighted imaging is often a time-consuming procedure as signal averaging is applied for improving the SNR. In a previous work we proposed a novel framework where, instead of signal averaging at very few b-values, 21 linearly spaced b-values over a wider range are measured without averaging [1]. In this context a model-based iterative algorithm was applied to estimate and to correct for Rician bias [2], resulting in more robust ADC maps. However, the proposed framework relies on pixelwise non-linear regression leading to computation times of several hours for a single patient. As this is impractical in a clinical context, parameter estimation by means of deep learning can significantly decrease computation time and might be a more viable option [3,4]. Furthermore, noise reduction in DWI signal through deep learning-based reconstructions have shown potential in improving image quality [5-8].
Expanding on previous works, deep learning reconstructions based on convolutional neural network (CNN) architectures were considered here for denoising prostate DWI. This approach to deep learning-based reconstructions reduces computation time and achieves similar image quality to that of model-based techniques.
Informed consent of the patients was received prior to imaging [9]. A multi-b sequence was used to acquire diffusion-weighted (DW) images at 21 linearly distributed b-values, ranging from 0 to 2000 [s/mm²], for three orthogonal diffusion encoding directions. A self-supervised training was implemented, as depicted in Figure 1, together with a five-fold cross-validation. The acquired DWIs, excluding the b0-image due to perfusion effects, were used both as input and the target for the neural network (NN). The resulting output of the NN are parameter maps depending on the fitting model used for denoising. Here, the kurtosis model S(b) = S0*exp(-bD+b²D²K/6) was considered to describe the signal. Subsequently, Rician bias was added by using a Gaussian noise map derived from OBSIDIAN [2]. The output images were then compared with the input using an L1 loss function. During inference the denoised DWIs from the fitting model were used as the output, i.e. no noise map from OBSIDIAN is necessary. In this study, standard UNet architecture [10], Attention based UNet [11] and Residual Attention UNet [12], were evaluated. The comparison of DW images from the clinical protocol, OBSIDIAN and the network models is shown in Figure 2. The images appear similar at low b-values but differences are observed at higher b-values, with images from OBSIDIAN and the NNs having lower intensity compared to the clinical images. Some anatomical structure seen in clinical images at b= 100 [s/mm²] vanishes at higher b-values, but to a lesser extent in OBSIDIAN and the different NNs.
As seen in Figure 3, the overall appearance of the ADC maps from the NNs are similar to OBSIDIAN with preserved anatomical structures and intensity distribution. ADC maps of the network models and OBSIDIAN, in comparison to raw- and clinically derived ADC maps, appear brighter in all areas as a result of performing bias correction. There were only subtle differences in the overall appearance between the NNs.
The estimated ADC values in Figure 4 by the network models are higher than clinical ADC and similar to OBSIDIAN. The ADC estimated by Residual Attention UNet were most comparable to that of OBSIDIAN. Overall, all employed models yielded reasonable estimates with varying degrees of uncertainty and ADC values across different network models. The visual quality of NN-generated DW images is very similar to the model-based algorithm OBSIDIAN, and visually more appealing compared to clinical DW images because of the lower signal intensity outside the prostate due to Rician bias reduction. With ADC maps no significant improvement in visual quality was observed. However, with ADC estimations both denoising methods result in higher estimates than clinical ADC as a result of bias reduction.
In this study DW image data was acquired from 25 patients, with test- and training data from 2 and 23 patients respectively. The size of the dataset constitutes a limitation and a larger dataset can be expected to improve the NN-based denoising. Using a UNet model in denoising for a single patient takes ~10 s with a GeForce GT 1030, which is a significant advantage over model-based algorithms and viable in clinical practice. Before total clinical integration, the method can be improved by including the noise information in the input data, and by using scanner-derived noise maps instead. Training with a larger dataset is also needed for robust performance. In the future, other model functions can be considered and more advanced NN-models, such as vision transformers (ViT), can be explored. In conclusion, NN-based denoising of DW image data has potential in clinical use and in achieving similar image quality to model-based algorithm with drastically reduced computation time.
Mustafa ABBAS (Gothenburg, Sweden), Fredrik LANGKILDE, Stephan MAIER, Stefan KUCZERA
15:30 - 17:00
#47385 - PG309 Optimized Lightweight Unsupervised Conditional GAN with Perceptual Loss Based MRI Reconstruction.
PG309 Optimized Lightweight Unsupervised Conditional GAN with Perceptual Loss Based MRI Reconstruction.
MRI offers good soft tissue contrast but long scan times[1].Traditional methods like Parallel Imaging (PI)[4] and Compressed Sensing (CS)[5] have improved reconstruction, but Deep Learning(DL) with Convolutional Neural Networks(CNNs) [8] offers superior results with fewer artifacts.However, DL faces challenges, particularly in applications like dynamic contrast enhancement and cardiac cine, where the extensive acquisition of fully-sampled data for training is time-consuming and impractical[6].
Recent studies have explored GANs for unsupervised MRI reconstruction.UC-GAN[11] allows training with limited data, avoiding the need for fully-sampled ground truth.It combines the Iterative Shrinkage-Thresholding Algorithm(ISTA)[14] with a data consistency step to handle complex data. To improve training stability, variants like WGAN and WGAN-GP add refined loss functions[13].However, UC-GAN and similar models face limitations, including high computational cost, unstable training, and poor perceptual features.To address these, we propose OLP-UC-GAN:a lightweight UC-GAN that uses Depthwise Separable Convolutions(DWSConv) [16] to reduce computation and Perceptual Loss[17] to enhance visual quality. It learns directly from under-sampled k-space data, improving both reconstruction accuracy and efficiency for better clinical utility.
We propose OLP-UC-GAN for MRI reconstruction, integrating DWSConv[16] into the generator of UC-GAN[11] to enable a lightweight architecture.The generator is based on ISTA[14]; and a Perceptual Loss from a pre-trained VGG-19[17].Proposed model discriminator employs a CNN [8] with residual connections and Leaky ReLU activations to ensure stable gradient flow.
Proposed model generator receives under-sampled complex k-space data as input and provides the reconstructed 2D complex-valued image.A data consistency step is also applied on the generated image to align the reconstructed image with the acquired k-space measurements by transforming it back to k-space, modulating with coil sensitivity maps, and masking. The discriminator then distinguishes between the real and reconstructed k-space measurements.
Training and evaluation are conducted on complex-valued 3T human knee MRI data available online[12], acquired with an 8-channel coil and a 3D FSE sequence. ESPIRiT is used for coil sensitivity estimation[15], and the data is under-sampled retrospectively with a uniform density mask at an acceleration factor of R=4.
In the context of MRI reconstruction, the multi-channel Fourier acquisition can be written as (Eq. 1)[11] : x = Bz + ɛ where x is the k-space data, B is the imaging model (including subsampling, Fourier transform, and coil sensitivity modulation), z is the reconstructed image, and ɛ is complex Gaussian noise. B is sampled from a known distribution p_B, leading to k-space data distribution pₓ.The conditional GAN objective is[11] :min┬G max┬D E_(x~p_(x ) ) [q (D(x))]+ E_(x~p_(x ),B~p_B ) [ q (1- D(B (G(x))))]( Eq.2)Here, G is the generator, D the discriminator, and q(v) = v denotes the WGAN-GP loss [13], promoting stable training. Additionally, a Perceptual Loss [17] based on VGG-19 features is added to ensure visual similarity between G(x) and the ground truth, improving high-level feature fidelity. The final objective thus combines adversarial and perceptual components to enhance reconstruction quality. Figure3 illustrates that the proposed method significantly improves knee MR image reconstruction over CS [5] and UC-GAN [11].Table1 presents quantitative results using PSNR, NRMSE, and SSIM.The proposed method achieves the highest PSNR (28.68 vs. 20.89 for UC-GAN and 18.75 for CS), the lowest NRMSE (0.26 vs. 0.38 and 0.75), and the highest SSIM (0.94 vs. 0.88 and 0.68), indicating superior reconstruction accuracy and structural fidelity. The proposed OLP-UC-GAN model introduces key enhancements that significantly improve MR image reconstruction.Integration of DWSConv[16] into the generator architecture results in better preservation of fine structural details and higher quantitative metrics, while also reducing model parameters for improved computational efficiency, which is critical for enabling faster processing and deployment in resource-constrained or real-time clinical environments.Additionally, incorporating Perceptual Loss[17] into the objective function aligns the reconstructions more closely with high-level features of the ground truth,enhancing visual fidelity.These modifications collectively demonstrate the effectiveness of the proposed method in achieving accurate and efficient reconstruction. The proposed method combines DWSConv and perceptual loss to enhance both computational efficiency and image quality.Results show improved MRI reconstruction across key metrics, highlighting the value of advanced convolutional techniques in deep learning-based medical imaging.This approach ensures accurate reconstruction while preserving structure and reducing computational cost.
Mahrukh MAZHAR (RAWALPINDI, Pakistan), Dr. Faisal NAJEEB, Dr. Hammad OMER
15:30 - 17:00
#46622 - PG310 Advanced Reconstruction for Arterial Spin Labeling MRI with Radial Sampling.
PG310 Advanced Reconstruction for Arterial Spin Labeling MRI with Radial Sampling.
Arterial spin labeling (ASL) is a promising non-invasive MRI technique for perfusion quantification, but it still faces the challenges of low signal-to-noise ratio (SNR) and relatively long scan times. This is especially true for multi-pulse-delay measurements to capture temporal regional variations of the magnetically labeled blood bolus. Typical ASL scans are based on Cartesian data acquisition, where the data generated are assigned to a time point (k0). To obtain time-resolved information, the scan must be repeated at the new time point, resulting in proportionally longer acquisition times. In contrast, radial sampling allows continuous acquisition of perfusion-weighted images (PWIs) with multiple post-labeling delays (PLDs) within a single scan [1]. Expected advantages are higher time efficiency, higher subsampling, and robustness to motion artifacts. Limitations of continuous radial acquisition are the signal attenuation of the labeled blood bolus and the still low contrast-to-noise ratio (CNR). To enhance image quality, particularly at later PLDs, image reconstruction is performed using the Berkeley Advanced Reconstruction Toolbox (BART) [2] with total generalized variation (TGV) [3] and infimal convolution of total generalized variation (ICTGV) [4] regularization, incorporating ASL-specific regularization on the PWIs [5].
A radial FLASH sequence was extended with a pseudo-continuous arterial spin labeling (PCASL) [6, 7] protocol (see Fig. 1). Sampling followed a golden-angle pattern [8], using a 5° flip angle and TR = 2.5 ms. The golden angle was incremented across averages while maintaining the same spoke pattern for each control/label pair. Two-dimensional ASL brain measurements were conducted on a 3T scanner (Siemens Healthineers, Erlangen, Germany) in a healthy volunteer. For continuous measurements, 260 spokes were acquired per average with a labeling duration (LD) of 1800 ms and a post-labeling delay (PLD) of 800 ms. To evaluate the signal quality of labeled blood flow from the continuous measurement, single-delay (SD) images with 65 spokes were acquired as ground truth for comparison. A total of 20 averages were acquired for each measurement. For image reconstruction k-space data was segmented into four frames, with each frame reconstructed from 65 consecutive spokes. Images were reconstructed using the parallel imaging compressed sensing (PICS) [9] tool from BART, applying ASL-TGV or ASL-ICTGV regularization with a spatio-temporal constraint on the PWI [5]. This constraint enables joint emphasis on spatial and temporal features, which is beneficial for the temporal dynamics critical to ASL imaging. PWIs were averaged over 20 repetitions. Reference reconstructions without (ASL-specific) regularization were performed. Additionally, the same k-space dataset was highly undersampled by dividing it into 12 frames, each reconstructed from 21 consecutive spokes. Fig. 2 compares continuously acquired PWIs reconstructed with different regularization methods (no regularization, TGV, ASL-TGV, ASL-ICTGV) to reference SD images. PWIs at later PLDs appear more detailed and structurally similar to their SD reference when ASL-specific reconstruction is performed. In contrast, without ASL-specific reconstruction, PWIs at later time points become increasingly noisy. Fig. 3 shows the reconstructed PWIs from the highly undersampled k-space dataset. Similar improvements are observed as in Fig. 2, but the effects of strong undersampling further highlight the advantages of ASL-specific regularization. The ASL-TGV and ASL-ICTGV reconstructions yield PWIs of reasonable quality for later time points, whereas the corresponding TGV results are nearly indistinguishable from noise. No visible differences were observed between ASL-TGV and ASL-ICTGV regularization. The radial readout scheme enables continuous perfusion signal acquisition within a single scan, which may be beneficial for kinetic model fitting to estimate physiological parameters. This approach may also enhance robustness to motion, potentially improving reliability in challenging subjects. The integration of TGV/ICTGV regularization combined with an ASL-specific regularization on the PWI in BART, facilitates the reconstruction of continuously acquired radial ASL data with improved image quality, particularly at later PLDs. These advanced regularization strategies are especially valuable in ASL due to its inherently low SNR. We successfully combined continuous radial ASL data with ASL-specific TGV and ICTGV regularization, resulting in improved image quality at later post-labeling delays. Future work will focus on extending this method to 3D for whole-brain coverage, enhancing labeling efficiency through background suppression, and comparing its performance to conventional Cartesian ASL sequences.
Viktoria BUCHEGGER (Graz, Austria), Ingrid BARTH, Ingmar Sören SORGENFREI, Philip SCHATEN, Rudolf STOLLBERGER, Martin UECKER
15:30 - 17:00
#47605 - PG311 BART Streams: End-to-End Latency of Real-Time MRI.
PG311 BART Streams: End-to-End Latency of Real-Time MRI.
The Berkeley Advanced Reconstruction Toolbox [1] is an MRI reconstruction framework and recently received support for
streaming and real-time reconstruction [2].
Real-time reconstruction is especially useful when directly connected to the MRI scanner,
as this enables e.g. live-streaming MRI images from the body, which can improve interventional MRI.
In this work, we describe a method for measuring end-to-end latency of
real-time MRI systems and apply it to an iterative real-time reconstruction method [3]
implemented in BART.
BART contains a set of tools for reconstruction and data/image processing.
Typically, several tools are successively applied to the data, usually with a reconstruction script written e.g. in BASH or Python.
Previously, each tool would wait until the previous steps were finished.
Recently, we added the options to stream data and to let each tool operate iteratively on subsets of the full data [2].
These changes enable real-time reconstruction.
Fig. 1 gives a graphical overview of a simple real-time reconstruction pipeline for Cartesian data.
We propose an improved [4] method for analyzing the latency of real-time MRI systems, which is described in Fig. 2.
Using a long, stiff string (fishing line), a movement is created in the scanner from the control room.
This is recorded using a camera both directly by observing a marker attached to the string, and through the MR images shown on the MRI console.
A Fairphone 4 (Fairphone B.V., Amsterdam) is used as the camera, which supports a framerate of 120fps.
This is considerably higher than the expected real-time MRI framerate and should therefore provide sufficient accuracy for the latency measurements.
The video is split into two image series, segmented, and the resulting apparent object speed is calculated.
The velocity curves are cross-correlated to obtain the total delay between the marker movement and the movement displayed on the MRI console.
This delay represents the end-to-end latency, i.e. the full delay introduced by all components of the real-time MRI system.
Measurements were performed on a 3T scanner (Siemens Healthineers, Erlangen), using a radial FLASH sequence with 9 spokes, base resolution 128, TE=1.09ms and TR=1.75ms.
The data is streamed to and from the scanner directly in BART streams format, reconstructed using real-time NLINV [3] and subjected to a five-frame median filter. The latency measurement methodology is validated by assessing the delay between the moving object inside and outside of the scanner, without MRI, i.e. when the camera simultaneously optically observes both ends of the string.
We found a delay of 0 frames, confirming that there is no additional offset within measurement accuracy.
Fig. 3 shows a snapshot from the video which was processed to determine the latencies. The MR image of the suspended test tube and the marker are clearly visible.
Results from the latency measurements are shown in Fig. 4.
Initially, there is a high latency of several seconds.
The latency decreases and reaches a steady state of approximately 200 ms after less than 3 seconds.
This represents an end-to-end latency - the reconstruction time per frame, as determined from timestamps, is much lower (~ 40 ms). The latency measurement shows that BART reconstruction scripts can achieve good latencies.
Time needed for reconstruction is only a fraction of the total latency.
Possible reasons for the initially high latency include initialization of GPU memory, starting of processes etc..
One contributing factor to the additional delay observed in the end-to-end latency is the response of the median filter.
This explains three frames or about 47.25 ms of the delay. The origin of the remaining latency is currently unclear.
Some possible reasons include our custom software for interfacing BART and the scanner, or data processing on the MRI.
Concerning the latency measurement methodology, the setup is much simpler compared to our previous method [4]. A simple latency measurement setup was devised and tested, with which we quantified the real-time quality of the BART streaming reconstruction.
The time required for the reconstruction step was found to be small compared to the end-to-end latency.
Philip SCHATEN (Graz, Austria), Moritz BLUMENTHAL, Martin UECKER
15:30 - 17:00
#47747 - PG312 Addressing Motion Outliers in Four-Dimensional Cardiac Flow MRI Reconstruction using Outlier Rejection.
PG312 Addressing Motion Outliers in Four-Dimensional Cardiac Flow MRI Reconstruction using Outlier Rejection.
4D flow MRI captures and quantifies blood flow dynamics in three-dimensional space over time. Utilizing k-space segmentation, the data collection over multiple cardiac cycles requires managing substantial 4D data along the spatial and temporal dimensions. Heart rate variability and respiratory motion further complicate the data acquisition process by introducing outliers that can degrade image quality hindering accurate flow visualization. To address these challenges, a range of standard navigator gating and respiration control techniques are utilized [1]. However, these techniques remain demanding and often fail to fully eliminate the motion-induced outliers leading to image degradation. Advanced imaging methods like Compressed Sensing (CS) can also be utilized for 4D Flow MRI reconstruction that allows for sparse and random data acquisition, followed by nonlinear data recovery, maintaining image quality in the reconstruction process [2]. Although CS methods outperform the prior, non-sparsity based techniques, such as SENSE and GRAPPA, they rely on the model [2], which may not hold in case of uncompensated motion. Another approach to handle motion-induced outliers in the collected k-space data is Compressive Sensing with Outlier Rejection (CORe) [3], recently proposed in literature that explicitly models the outliers as additive variables, facilitating the separation of the measured data into outlier estimates and the k-space data for reconstruction [3]. However, this model does not capture outliers efficiently because of the varying types of outliers present in an individual 4D flow dataset due to physiological movements and fluctuating blood. This paper proposes, a new method, which further enhances CORe by differentiating the range and extreme outliers in the k-space data for physiological movements. The efficacy of the proposed method is validated by performing reconstruction on 4D cardiac dataset for the mitigation of motion outliers and was also compared with the conventional CS and CORe reconstructions.
The key steps of proposed methodology are shown in Figure 1. The process starts with 4D cardiac dataset preprocessing through parameter initialization i.e., acceleration rate, number of iterations, regularization parameters etc. as shown in Figure 1. Haar-Wavelet transform was utilized (because of its simple domain operations) to improve reconstruction quality by enforcing sparsity in the wavelet domain. The proposed method is outlined in Equaion (1):
x = (1 / σ²) * ‖Ax - y + v₁ + v₂‖₂² + R(x) + λ₂ * ‖g(v₁)‖₁ + λ₃ * ‖g(v₂)‖₁ (1)
Where Ax - y is the Fidelity term, R(x) is the Wavelet transform and introduction of variables v₁ and v₂ are used to model the outliers within the dataset range and outside the range. By taking the L₁ norm of the functions g(v₁) and g(v₂) which denote the group of outliers, group sparsity is promoted in the k-space. These sparse outliers are classified into range and extreme outliers by utilizing IQR- Inter Quartile Range method, and a random slice containing outliers is displayed in Figure 2. The outliers are later removed by empirically setting different threshold values during suppression process.
For reconstruction, the proposed method employs Split Bregman algorithm [5] as shown in Figure 1. It was chosen for its efficiency in computing sub-problems iteratively by separating data fidelity, sparsity, and outlier’s term, solving them using soft thresholding with Bregman updates. This enables robust and fast convergence of the proposed method. The resulting Four-Dimensional MR images were then reconstructed and post processed to visualize blood flow in the aorta, after background phase correction, adjusting the phase inconsistencies. Experiments were performed on prospectively acquired 4D flow data of the ascending aorta and is openly available [4]. Figure 2 and Figure 3 highlight the 4D reconstruction results including flow visualization and mean velocity outputs in Aorta, respectively. The proposed method outperforms CS [2] and CORe [3] methods, in terms of PSNR and SSIM values as given in Table 1. While velocity graphs for all the methods might suggest similarities, a more rigorous assessment was performed via Pearson correlation coefficient [6] and R² [7] values between the velocity magnitude image and the ground truth, where proposed method shows better accuracy in terms of reconstructed image magnitude and velocities, thus contributing to a clearer and more interpretable depiction of cardiac flow. This paper introduces a novel reconstruction method to mitigate the motion outliers in 4D flow MRI and compares its performance with conventional CS and CORe methods. The results from 4D Cardiac dataset show that the proposed method is more effective in suppressing motion outliers while preserving high image quality, making it a superior approach for accurate 4D flow MRI reconstruction.
Muhammad SHARJEEL, Umair KHALIL (Islamabad, Pakistan), Omair INAM, Hammad OMER
15:30 - 17:00
#45419 - PG313 Introducing gammaSTAR Reconstructions: Demonstration of Vendor Neutral MR Data Acquisition and Reconstruction on Tabletop Systems, MR Simulators and 3T Systems.
PG313 Introducing gammaSTAR Reconstructions: Demonstration of Vendor Neutral MR Data Acquisition and Reconstruction on Tabletop Systems, MR Simulators and 3T Systems.
Magnetic resonance imaging (MRI) is one of the most versatile imaging techniques in modern clinical settings. Besides high-field and ultra-high field applications, MRI has advanced to low-field configurations, enabling imaging of small probes using tabletop systems. Finally, educational purposes benefit from the recent development of fast MRI simulators [1]. The gammaSTAR sequence development framework offers a versatile tool to implement vendor-agnostic MR sequences to control the mentioned variety of MR systems [REF]. However, to date, image reconstruction was still performed using the vendor-specific reconstruction pipelines, which can be a problem for quantitative MRI tasks or the development of imaging biomarkers as this might introduce variabilities into the image quality on different systems. In this work, we introduce gammaSTAR reconstructions which overcome the current limitation by implementing a fully functional reconstruction server, connected to the gammaSTAR framework. With this, we demonstrate vendor-agnostic MR imaging on a 3T Siemens VidaFit system (Siemens Healthineers, Erlangen), on an Ilumr tabletop system (Resonint, Wellington) and using the MRZero simulation tool.
All gammaSTAR sequences utilize the ISMRMRD standard [3] to assign relevant measurement flags to individual data readout events. This allows to implement a highly flexible reconstruction server, which applies the same series of reconstruction modules for all non-Cartesian (using mri-nufft toolbox [4]) and Cartesian acquisitions (cf. Fig 1a). Whether a module needs to be applied is decided based on the received data dimensions and ISMRMRD flags. The reconstruction server is provided as an OpenRecon compatible Docker image. The complete workflow of measuring/simulating data is demonstrated in Fig. 1b. MR sequences can be implemented in a modular fashion using the gammaSTAR sequence frontend. From the frontend, the corresponding sequence protocol can also be adjusted. The adjusted sequence is then sent to a variety of systems using specific drivers. Finally, resulting image data is displayed in the frontend. To validate the flexibility of the reconstruction tool, a large set of different two- and three dimensional sequences is investigated. Using MRZero 2D RARE, 2D RADIAL, 2D FLASH, 2D bSSFP and 2D Spin-Echo EPI sequences were simulated. On the Siemens VidaFit 3T system, 2D RARE, 3D MP-RAGE and 2D EPI sequences were applied. Finally, 2D RARE, 2D FLASH and 2D Spin-Echo EPI sequences were tested on the Resonint Ilumr system. Protocols were adjusted to cover a large range of available options such as golden-angle schemes for the radial acquisition, parallel imaging using external and integrated reference scans as well as partial Fourier setups. Fig. 2 shows results from simulations with the MRZero simulator and a large variety of contrasts and acquisition strategies. Fig. 3 shows results from the Siemens VidaFit 3T full body MR system using parallel imaging strategies. Fig. 4 shows finally shows results from the Resonint Ilumr system. MR images were successfully reconstructed using gammaSTAR reconstructions for the Siemens VidaFit 3T system, the Resonint Ilumr system and the MRZero simulator. Applied sequences and protocols covered a large range of possible configurations. This demonstrates the flexibility of the reconstruction server in combination with the gammaSTAR sequence development framework, offering the user the option to test sequences using the MRZero simulator before executing them on real full body or tabletop MR systems. Our solution especially benefits from the close link between the sequence development and respective reconstruction tools. This represents an advantage over other open source solutions such as Gadgetron [4, 5] which aim to support a broad range of sequence frameworks, making it more difficult to maintain a common standard. The reduced quality of partial Fourier reconstructions (cf. Fig. 3) as well as the low signal-to-noise ratio on tabletop systems (cf. Fig. 4) will be improved in the future. In addition, image normalization using a body coil reference will be included. Finally, the reconstruction server will be extended by image processing steps, such as automatic quality assessment (AQUA) and quantification options such as generation of T1/T2 maps or calculation of cerebral blood flow maps in case of Arterial Spin Labeling experiments. We demonstrated the vendor-independent gammaSTAR reconstruction server in combination with the gammaSTAR sequence development tool. The flexibility of the reconstruction server was verified in three different scenarios: A full body Siemens VidaFit 3T system, a low-field Resonint Ilumr tabletop system and the virtual MRZero simulator. Future work will further extend the compatibility to various other systems. In addition, a fully virtual MR workspace will be investigated which allows direct transition of developed sequences and reconstruction techniques to real hardware.
Jörn HUBER (Bremen, Germany), Snawar HUSSAIN, Vincent KUHLEN, Arne NEISSER, Lukas SCHENK, Simon KONSTANDIN, Matthias GÜNTHER, Mariia KLIMENKO, Daniel HOINKISS
15:30 - 17:00
#47801 - PG314 Accelerating 4D flow MRI construction using FPGA: FPGA based accelerator for rapid 4D Flow MRI reconstruction.
PG314 Accelerating 4D flow MRI construction using FPGA: FPGA based accelerator for rapid 4D Flow MRI reconstruction.
4D flow MRI is an advanced imaging technique for visualizing blood flow dynamics. It plays a critical role in the early and accurate diagnosis of cardiovascular conditions such as congenital heart disease (CHD), aortic wall disease, valvular heart disease, pulmonary hypertension, and peripheral artery disease. This technique involves acquiring one reference image and three velocity-encoded images along orthogonal directions across multiple cardiac cycles [1]. Consequently, the k-space data spans six dimensions: frequency encoding, phase encoding, slice selection, coil channels, temporal frames, and velocity direction. Such high-dimensional data acquisition results in computationally intensive and time-consuming image reconstruction, which poses a barrier to its widespread clinical adoption.
To address this challenge, this study presents a novel FPGA-based hardware accelerator architecture for compressed sensing reconstruction of 4D Flow MRI data. The proposed system leverages the parallel processing capabilities of the Xilinx Zynq-7000 SoC to significantly reduce reconstruction time without compromising image quality.
The architecture, illustrated in Fig. 1a, integrates multiple FPGA based parallel computational blocks to accelerate large-scale complex matrix-matrix multiplications. In the proposed accelerator design, the DMA (Direct Memory Access) facilitates high-throughput data transfer between the accelerator and DDR3 memory via the AXI-Stream interface, minimizing CPU intervention and significantly reducing processor overhead [2]. CPU (ARM core of zynq SoC) functions as memory controller and host for arbitration module in its program memory [3]. The arbitrator is responsible for partitioning data arrays coordinating DMA transfer to enable efficient communication with parallel accelerators, ensuring fast and reliable data movement while maintaining consistency with original dataset.
Fig. 1b illustrates the design flow of hardware accelerator to compute the wavelet transform, which serves as a sparsifying basis in the regularization term of the compressed sensing objective [4] and is involved in the gradient computation during reconstruction [5]. As shown in Fig. 2, gradient computation dominates the overall reconstruction process, accounting for approximately 72% of the total reconstruction time High-level-synthesis (HLS) is adapted for designing hardware accelerator, targeting wavelet transform module used in compressed sensing. The algorithm, implemented in C/C++ is optimized using HLS compiler Directives i.e. HLS pipeline, HLS unroll to exploit fine-grained parallelism and maximizing throughput [6].
Design constraints are enforced to meet latency, and area targets suitable for the Xilinx Zynq-7000 platform, achieving a balance between performance and hardware resource utilization. Functional correctness is validated through C/RTL simulation and co-simulation, ensuring bit-accurate correspondence between the high-level model and synthesized RTL. Reconstruction time comparison between the proposed method and CPU-based implementation is presented in Fig. 3a. The results show that the proposed method outperforms the CPU-based compressed sensing reconstruction, achieving a fourfold acceleration in overall reconstruction time while maintaining image quality. Fig. 3b presents a comparison of 4D Flow MRI reconstructed images (magnitude and three velocity-encoding directions) for a single frame using both the CPU and the proposed method. The results demonstrate that the proposed method achieves image quality comparable to that of the CPU-based reconstruction. This research introduces a novel FPGA based SoC architecture for accelerating wavelet transform in sparsity gradient of compressed sensing for 4D flow MRI reconstruction. The results using 30-coils, 20-frames in-vivo 4D stress cardiac dataset shows that proposed method can achieve four-times faster reconstruction without compromising the reconstruction accuracy and making 4D flow MRI suitable for clinical adaptation.
Mutahar KHADIM, Hammad OMER, Omair INAM, Abdul BASIT, Muhammad ZUHAIR (Islamabad, Pakistan)
15:30 - 17:00
#47321 - PG315 Physics-Informed Temperature Field Reconstruction in Thermal Ablation Validated Using MRI Thermometry.
PG315 Physics-Informed Temperature Field Reconstruction in Thermal Ablation Validated Using MRI Thermometry.
Thermal ablation is widely used as a first-line treatment for malignant tumors [1–3], relying on extreme temperatures to destroy tumor tissue. Magnetic Resonance Imaging (MRI) thermometry enables real-time monitoring of temperature changes during procedures [4–6]. However, its precision is limited by measurement uncertainty, reduced accuracy near the applicator due to susceptibility artefact and partial volume effect [7,8]. To address these limitations, numerical heat transfer simulations can complement MRI data by improving spatial resolution and reconstructing temperature information lost due to artifacts or low signal quality [9–12].
Model description: A hybrid framework was developed to simulate temperature distributions during MRI-guided thermal ablation. The model relies on the solution of the heat equation in spherical coordinates, solved using the Laplace transform and finite difference methods. For each energy, a source term was specified as follows: For Laser-Induced Thermal Therapy (LITT), heat deposition is modelled as: Q(r,t)=Q0(t)exp(−βr). For Microwave Ablation (MWA): Q(r,t)=Q0(t)exp(−β√r). For Radiofrequency Ablation (RFA): (Q(r,t)=Q0(t)H(r−r0)), where H is the Heaviside function. A spherical temperature distribution is computed and mapped onto a 1-mm grid. This half-sphere is then extended into a cylinder to represent applicators with axial symmetry. The resulting 3D structure is aligned with MRI images using rigid registration. This workflow is illustrated in Figure 1. These solutions are then mapped onto a 3D voxel grid by averaging the temperature inside each voxel, aligned with the experimental MRI data, and a mask is applied to keep only reliable voxels. The model then estimates the heat source parameters Q0(t), β, and r0 by minimizing the voxel-wise error between simulated and measured temperatures over the entire ablation time. The model focuses on estimating heat source parameters while assuming constant thermal properties, simplifying the calculations without losing accuracy. Lastly a super-resolved, three-dimensional temperature field at a finer resolution than the MRI voxel size is reconstructed.
Device and experiments:
MWA was performed in bovine liver using a 14G antenna (AveCure, MedWave, USA) under MRI guidance. Ablation lasted 7 min 30 s, targeting 60 °C. A 1.5 T scanner (Magnetom Sola Fit, Siemens) acquired 13 slices every 4 s with a multi-shot EPI sequence (TE = 19 ms, TR = 47.3 ms, FA = 90°, matrix = 128×128, slice thickness = 3 mm, gap = 1.5 mm, FOV = 300×300 mm², BW = 953 Hz/pixel).
LITT was tested in 3% agar gel using a 976 nm diode laser (8 W, 1 min) coupled to a 240 μm diffuser (ALPhANOV, France). MRI (1.5 T Avanto, Siemens) monitored the ablation with a single-shot EPI sequence (8 slices, every 2 s; TE = 21 ms, TR = 2000 ms, FA = 40°, matrix = 128×128, thickness = 3 mm, FOV = 158×158 mm², BW = 1445 Hz/pixel).
RFA used an MR-compatible catheter (Vision-MR, Imricor) with two microcoils for tracking. Power levels of 5, 10, 15, and 20 W were applied in agar gel for 30 s. MRI thermometry (1.5 T Aera, Siemens) acquired 8 slices every 1 s with a multi-shot EPI (TE = 28 ms, TR = 930 ms, FA = 60°, matrix = 148×148, thickness = 3 mm, gap = 0.3 mm, FOV = 170×170 mm², BW = 1165 Hz/pixel).
Acquisition: For all acquisitions, temperature maps were computed using the Proton Resonance Frequency Shift after B0 drift correction. For each case, simulated and experimental temperature maps were compared, and the model successfully predicted temperature distributions that matched the experimental data. Each simulation step took less than 45 ms, and data alignment with MRI measurements averaged 87 s. Figures 2, 3, and 4 present temperature comparisons for MWA, LITT, and RFA, using a 5×5 voxel kernel centered near the applicator. The normalized root mean squared error reached up to 20 °C for MWA, 3 °C for LITT, and 7 °C for RFA. Higher errors in MWA are attributed to complex near-field effects not captured by the simplified model. In contrast, the compact symmetry of the laser diffuser in LITT yielded highly accurate predictions. This framework combines analytical modeling, numerical methods, and MRI thermometry to reconstruct thermal fields with improved resolution. By adapting the source term to each ablation technique, the model remains flexible and computationally efficient ( under 2 min total runtime). It recovers temperature in regions affected by artifacts and can predict the maximum temperature at the vicinity of the device without partial volume effect. Future work will integrate perfusion effects and apply the model to in vivo datasets for clinical validation.
Mariana DE MELO ANTUNES (Bordeaux), Ida BURGERS, Valery OZENNE, Luigi NARDONE, Max SEIDENSTICKER, Olaf DIETRICH, Manon DESCLIDES, Bruno QUESSON, Andrzej KUSIAK, Sandro Metrevelle MARCONDES DE LIMA E SILVA, Jean-Luc BATTAGLIA
15:30 - 17:00
#47604 - PG316 General reconstruction of parallel-transmit array B1+ profiles with enhanced noise robustness.
PG316 General reconstruction of parallel-transmit array B1+ profiles with enhanced noise robustness.
Parallel transmission (pTx) is essential in ultra-high field MRI to improve signal and contrast uniformity [1], but requires accurate measurement of the channel transmission profiles (B₁⁺) [2]. The interferometric pre-saturated turbo-FLASH (SatTFL) sequence [3-5] offers a valuable technique for this purpose, but its reliability deteriorates with a large B₁⁺ dynamic range of the combine, typically, circularly polarized (CP), mode [6,7]. In this case, a conventional B₁⁺ reconstruction method can drastically amplify noise propagation and impair subsequent pulse design. This problem was addressed recently in [8] by tailoring the interferometric scheme to the RF coil. However, this may appear insufficient if the dynamic range (max/min) of the interferometric modes (IMs) remains too high. In this work, we consolidate the SatTFL method by introducing a least-squares-based (LS) B₁⁺ reconstruction offering a flexibility in interferometric scheme and minimizing noise propagation.
The satTFL signal [9] depends on the saturation (α) and readout (β) flip angles (FAs), determined by complex Nch-dimensional B₁⁺ profiles (Nch is the channel number) and the corresponding IM matrices A and B (Eq. 1, Fig. 1A). Estimating B₁⁺ from measured noisy data s₁, s₂, … represents an inverse problem, with A and B given, and B₁⁻ and m₀ as additional unknowns.
The standard B₁⁺ reconstruction uses the S-std scheme (Fig. 1B), with A and B of the form [X;0;0] and [X;X;Y], where X is a single-column and Y is a square "interferometric" matrix [8,10]. With S-std, B₁⁺ is typically derived using the noise-agnostic workflow M-std (Fig. 1E), which involves computing s₁/s₂, s₃/s₂, … (steps 1-3) followed by solving an “interferometric” linear system (step 4). In the presence of noise, this method can be locally instable for voxels with low s₂ signal or poorly conditioned Y.
To address this problem, we propose (i) a method (M-LS) to estimate B₁⁺ via least-squares fitting of the SatTFL signal to Eq. 1, and (ii) a method for optimizing the IMs.
(i) We factorize B₁⁺ as B₁⁺=σb₁⁺ with σ=||B₁⁺||, and use a two-step workflow (Fig. 1F): 1) estimate the phasor b₁⁺ from the subset of signals acquired with α=0 (a linear least-squares problem); 2) reinject b₁⁺ into the full signal model and estimate σ by gradient descent (a 1D nonlinear least squares).
(ii) M-LS is not limited to the S-std configuration but applies to the more general S-Nsat scheme shown in Fig. 1C, where Nsat images are acquired with α≠0. A and B then take the form [X;0] and [X;Y], where X (resp. Y) is the encoding matrix for the Nsat (resp. Nch) unitary IMs. Heuristically, the X and Y blocks are designed separately, based on a representative B₁⁺ distribution and the objective function in Fig. 1D, Eq. 2. This approach ensures that, for each voxel, at least one IM yields a FA close to the target. The condition number constraint guarantees that the inversion of Y is noise-robust.
We applied the proposed approach on a Magnetom 7T MRI using the 8Tx32Rx Nova head coil and a home-made 8Tx32Rx Avanti2 prototype [11], which has a markedly different geometry and a larger CP-mode dynamic range. EM simulations on a spherical agar phantom (SP) returned CVs of 0.29 and 0.45 for the CP mode of the Nova and Avanti2, respectively.
We computed S-std, S-2sat, and S-3sat based on simulated SP’s B₁⁺ profiles, scaling the IMs to obtain 〈α〉=60° and 〈β〉=4°. SatTFL signals in the SP were simulated according to Eq. 1 (Fig. 1A) using a complex Gaussian noise source n with std(n)/m₀=1e-3; then, M-LS and M-std (when appropriate) were applied. We repeated this procedure 100 times to derive the average relative B₁⁺ error map, ∆. For Avanti2, we additionally found the optimal 〈α〉 and 〈β〉 for S-3sat via exhaustive search, minimizing the average error 〈Δ〉. The IM schemes were then tested in simulation for a human head model (HM) [12]; the FA-optimized S-3sat was finally applied in vivo on one subject. The simulation results for Nova and Avanti2 are shown in Figs. 2 and 3. The optimal average FAs for S-3sat were <α>=62° and <β>=8°. In vivo B₁⁺ acquisition (Fig. 4) with Avanti2 took 2 min 20 s, with the M-LS reconstruction of ~30s. The proposed M-LS B₁⁺ reconstruction algorithm reduces noise propagation and supports a wider variety of schemes compared to M-std. For Avanti2, M-std applied to S-std is prone to large errors (〈Δ〉 = 6.7%). Using M-LS reduces 〈Δ〉 to 3.1%, but the S-3sat scheme is necessary to bring the error down to a level comparable to Nova (〈Δ〉=2.3%). Optimized FAs <α>=62° and <β>=8° further decrease 〈Δ〉 to 1.2%. The SP-based scheme remains applicable for the head (〈Δ〉 = 1.3% on HM), but a more optimized scheme could be established iteratively from a database of in vivo B₁⁺ maps. The proposed method improves the quality of the reconstructed B₁⁺ data required for pTx pulse design and can be applied to a wide range of transmit arrays architectures. It may also be valuable at higher magnetic fields and for body 7T MRI.
Natalia DUDYSHEVA (Gif-sur-Yvette), Franck MAUCONDUIT, Vincent GRAS
15:30 - 17:00
#47597 - PG317 Evaluation of accelerated whole-body diffusion weighted imaging with deep learning reconstruction in patients with metastatic prostate cancer.
PG317 Evaluation of accelerated whole-body diffusion weighted imaging with deep learning reconstruction in patients with metastatic prostate cancer.
Whole-body (WB) MRI, including diffusion-weighted imaging (DWI), is recommended in patients with metastatic prostate cancer [1]. WB-DWI is used for tumour detection and treatment response assessment [2-4], but long acquisition times limit patient comfort and compromise scanner throughput.
Deep learning (DL)-based reconstructions have the potential to aid the acceleration of MR imaging while preserving diagnostic image quality [5,6]. Prior studies show improved image quality in DWI with DL-based reconstruction (DL DWI) but lack cancer type-specific quantitative evaluations [7-14].
Our study aims to evaluate image quality and apparent diffusion coefficient (ADC) estimates using an accelerated DL WB-DWI acquisition compared with standard WB-DWI in patients with metastatic prostate cancer.
Two prostate cancer patient cohorts, scheduled for a clinical WB-MRI examination at any treatment stage, were scanned on a 1.5T MR scanner after verbal consent.
Patients underwent routine clinical WB-MRI of 5-stations, and research sequences were performed at the end of the examination.
Accelerated DL-DWI was adapted from the routine WB-DWI protocol at our centre, using fewer signal averages and higher parallel imaging acceleration (Table 1).
DL-DWI data were reconstructed inline using a prototype deep learning-based variational network [15] trained on healthy volunteer DWI data, alternating between data consistency and learned regularization steps as presented in [11].
Cohort 1 (n = 10, mean age 70 ± 7 years) compared image quality between standard WB-DWI and accelerated DL WB-DWI acquisitions across a 4-station (neck to thighs) WB-DWI acquisition. Two experienced radiologists blinded to the type of DWI sequence, scored image quality for b900, b900 maximum intensity projection (MIP) and ADC images on a 4-point Likert scale (described in Figure 1).
Cohort 2 (n = 20, mean age 75 ± 9 years) compared ADC values from both protocols on a radiologically active focal bone lesion (size ≥ 1cm) from a single station. Focal lesions were delineated on standard b900 images, and the volumes of interest (VOIs) were transferred to the corresponding ADC maps for each patient (IDL ADEPT, ICR, UK). The median and interquartile range (IQR) of ADC estimate from all fitted voxels in the VOI for each patient were measured.
Statistical comparisons used Wilcoxon signed-rank tests with Bonferroni correction to compare image quality scores, and Bland-Altman analysis to compare ADC estimates. Cohort 1
The qualitative radiological scoring did not find significant differences between the standard WB-DWI and the accelerated DL WB-DWI protocols (p > 0.0025), with most of the examinations rated as ‘good’ in any of the qualitative metrics considered for both protocols (Figure 1).
Exemplar b900, ADC and b900 MIP images acquired with standard WB-DWI and DL WB-DWI are shown in figure 2.
Cohort 2
The cohort average lesion volume was 16.7mm3(range: 2.4 – 95mm3). The cohort mean and standard deviation of the lesion ADCs were 121.9 ± 41.5 (*10-5 mm2/s) for the standard WB-DWI and 122.3 ± 43.8 (*10–5 mm2/s) for the accelerated DL WB-MRI.
Bland-Altman analysis (Figure 3) showed no significant difference in ADC values between the standard WB-DWI and the accelerated DL WB-DWI protocols (p = 0.93). Similarly, the IQR of the ADC estimates was not significantly different between the two protocols (p = 0.63). The qualitative assessments demonstrated that an accelerated DWI protocol with DL-based reconstruction can reduce acquisition time by 7min and 10s (or 37%) for a 5-station WB-DWI examination without reducing image quality. This result aligns with previous studies reporting maintained or superior image quality and SNR outcomes in high b-value and ADC images of DL-reconstructed DWI compared to a standard DWI protocol [7-9,11,14].
In our 20-patient cohort, accelerated DL DWI did not affect ADC quantification in bone lesions, consistent with prior studies on WB and spine imaging [8,14]. However, conflicting results have been reported for the breast and liver, with some studies finding no differences [10,12] or significantly higher ADC values [11] for DL DWI compared with standard DWI protocols.
Unlike other reports [14], the DL DWI sequence was mainly accelerated by reducing b-value averages, keeping the diffusion mode and scheme unchanged. This minimizes added eddy-current distortions, enables direct protocol comparison, and clarifies DL technology’s impact on ADC quantification
A limitation of this study is the relatively small cohort size and single-centre design, which may limit the generalisability of our findings. However, the study included patients with metastatic prostate cancer undergoing WB-MRI examinations at any treatment stage and is therefore representative of this WB-MRI patient population. An accelerated DWI protocol with DL-based reconstruction reduced the acquisition time of WB-DWI by 37% without compromising image quality or ADC estimates.
Evanthia KOUSI (London, United Kingdom), Mihaela RATA, Dow-Mu KOH, Christina MESSIOU, Nina TUNARIU, Georgina HOPKINSON, Emily EVANS, Omar DARWISH, Elisabeth WEILAND, Thomas BENKERT, Jessica WINFIELD
15:30 - 17:00
#46634 - PG318 MULTI-CONTRAST SUPER-RESOLUTION FOR MRI WITH A VARIATIONAL APPROACH: DISENTANGLING THE CHALLENGES.
PG318 MULTI-CONTRAST SUPER-RESOLUTION FOR MRI WITH A VARIATIONAL APPROACH: DISENTANGLING THE CHALLENGES.
Several medical protocols benefit from the wide variety of soft tissue contrast that can be obtained from MRI with high spatial resolution. Nevertheless, acquiring high-resolution 3D MRI volumes for different contrasts can require significant acquisition time. In this respect, multi-contrast reconstruction strategies offer a potential solution [1] by taking advantage of the redundancy of information present in the measurements. Techniques such as compressed sensing and super-resolution (SR) use the anatomical information from one specific contrast to reconstruct another under-sampled contrast [2]. In this work, we study the necessary and sufficient conditions for applying SR to multi-contrast reconstruction from a variational point of view. Quantitative and qualitative results on in vivo T1 and T2 weighted 3D TSE data at different stages are presented.
The 3D SR problem we consider is to reconstruct an isotropic volume X from an under-sampled one Y, so that
Y~HX, (1)
Where H is the forward operator that establishes the correspondence between the high-resolution object X and the low-resolution acquisition Y. It considers the geometric transformations, down-sampling and the PSF.
In addition, if X_Ω represents a reference isotropic volume, the inverse problem (1) can be posed as follows:
X ̃"\∈\ " 〖"Argmin" 〗_X 1/2 ‖Y-HX‖_2^2+βR(X,X_Ω ) (2)
Where R is a regularization term that contains the a priori information about the volume X ̃ to be reconstructed. In this work, We consider:
R(X,X_Ω )=‖DX- e_Ω DX‖_2^2 with 〖 e〗_Ω= 〖DX_Ω〗^2/(〖DX_Ω〗^2+ τ^2 ) (3)
Where D is the finite difference operator, e_Ω is an edge map obtained from X_Ω (fixed during optimization), and τ>0 a parameter which thresholds〖 DX〗_Ω. Gradients in (3) share the edge localization and direction [3], then the idea of e_Ω is to extract these features. Accordingly, the term (3) can be interpreted as an edge-weighted Tikhonov regularization. Given that e_Ω∈ [0,1] the smoothing in (3) is applied proportionally to the relative importance of a contour.
Experiments: First, reference isotropic volumes in vivo (1mm^3) were acquired using a 3D TSE sequence on a 3T MRI system (MAGNETOM Vida, Siemens Healthineers, Germany) with (TE, TR) = (11, 700) MS and (400, 3200) MS for T1-w and T2-w images. Then, from isotropic volumes, we simulated a down sampled anisotropic volume by using the direct operator H (1 x 1 x 4 mm^3). Then, an isotropic volume X ̃ is reconstructed by optimizing (2). Iterative methods were implemented to solve (2), with a relative cost of 10^(-7) order. Regularization parameter β was tuned to maximize the Peak-Signal to Noise Radio (PSNR) and the Structural Similarity Index Measure (SSIM) see Table 1. In order to study the influence of structure knowledge in reconstructions, four cases were considered:
(1) Ground truth contour map: Information about the same contrast is used, that is, if a T1-w volume is reconstructed, then 〖DX〗_(Ω_T1 )is used in (3).
(2) A mutual contour map:
〖 e〗_(Ω_(T1∩T2) )= (〖|DX〗_(Ω_T1 ) | 〖|DX〗_(Ω_T2 ) |)/(〖|DX〗_(Ω_T1 ) | 〖|DX〗_(Ω_T2 ) |+ τ^2 )
(3) Constraint contour map:〖 e〗_Ω contains only information from the other contrast, that is, if a T1-w volume is reconstructed, then 〖DX〗_(Ω_T2 )is used in (3).
And (4) A gradient projection P_(e_Ω ) proposed in [2] (〖 e〗_Ω contains only information from the other contrast) where
R(X,X_Ω )=‖DX- e_Ω ⟨e_Ω│DX⟩‖_2^2 with 〖 e〗_Ω= (DX_Ω)/√(‖DX_Ω ‖^2+ τ^2 ) (4) A global view for T1-w and T2-w images of an axial slice is showed in figure 1. Figure 2 shows a zoom of a coronal slice. In general, significant and consistent high-resolution content is recovered in HR images (blue arrows), as well as vertical artefacts due to sagittal under sampling (red arrows) are corrected. When a ground truth contour map is used fine details are recovered which is reflected in quantitative measurements (see Table 1). In a second stage, a mutual contour map test showed to be very important, since we found that informations about edge localization and direction are presents in the other contrast. However, when only information from other contrast contour map is considered, unrealistic intensity jumps artefacts are produced (yellow arrows). Hence, a complementary a priori about gradient information must be made, for example the gradient projection proposed in [2] allows for each gradient direction to include the information from the other directions, this attenuates the intensity jumps artefact without losing the gained structures. The experiments conducted in this study allow us to verify and demonstrate that a priori information about edge location, direction and weighting are the necessary and sufficient conditions for satisfactorily solve the SR problem. This work suggests that multi-contrast SR is feasible if a high-resolution contour map is available. Reconstruction methods that inherently provide edge information such as DMS [4], will be investigated in this regard.
Luis AMADOR (Lyon), Marion FOARE, Olivier BEUF, Hélène RATINEY, Eric VAN-REETH
|
Poster hall |
17:00 |
TIME FOR A BREAK
- Coffee and refreshments will be available at the cash bar.
|
17:30 |
"Thursday 09 October"
Added to your list of favorites
Deleted from your list of favorites
A17
17:30 - 18:30
HOT TOPIC DEBATE
Only Open Science is True Science
17:30 - 18:30
Opponent.
Klaas PRÜSSMANN (Professor) (Keynote Speaker, Zurich, Switzerland)
17:30 - 18:30
Proponent.
|
Auditorium 900 |
18:30 |
WELCOME RECEPTION
|
Auditorium 900 |
Friday 10 October |
08:30 |
"Friday 10 October"
Added to your list of favorites
Deleted from your list of favorites
A20
08:30 - 09:30
FT2 Oral - Translation: Fetal to Adult
FT2: Cycle of Translation
08:30 - 08:40
#45873 - PG016 Fetal MRI with a wearable coil vest at 3 T — proof of concept and perspectives.
PG016 Fetal MRI with a wearable coil vest at 3 T — proof of concept and perspectives.
Recently, fetal MRI has become the secondary imaging modality in clinical routine [1,2]. Despite high volumetric imaging quality and rich soft tissue contrast, it has limitations such as tedious patient positioning and the tradeoff between patient comfort, imaging speed, and quality. Besides the widely used 1.5 T MRI in clinical routine, low-field fetal MRI allows for more accessible MR exams [3] but high-field MRI at 3 T provides highest SNR and anatomical details [4]. Clinical fetal MRI is mostly performed using the integrated spine coil and a semi-flexible coil for abdominal or thoracic applications.
The concept of using flexible coils has already been proven to be beneficial for breast [5,6], knee [7,8], neck, ankle or spine [9], and pediatric MRI [10,11]. With pregnant patients, coil flexibility allows for adjustable patient positioning, either in supine or lying on the side depending on patient preferences and medical considerations. Moreover, dedicated wearable coils designed for the target application can optimize SNR and reduce measurement time, and consequently, allow for a comprehensive and patient-friendly fetal MRI exam.
In this work, the aim was to demonstrate the feasibility of high-resolution fetal 3 T MRI employing a wearable coil vest (“Bracoil” [5]) initially designed for breast MRI. To this end, we tested its usability on a phantom and in vivo, and derived implications for the development of a dedicated wearable fetal coil.
The MRI workflow in our study was to first assess the coil coverage and tune sequence parameters on an anatomy- and tissue-mimicking fetal phantom due to the restricted measurement time (<30 minutes) on pregnant volunteers. The in vivo study was IRB-approved (“EDEN”, Nancy, France, ClinicalTrials.gov Identifier: NCT05218460) and written informed consent was obtained from a volunteer (29 years, abdomen circumference 92 cm, 34 weeks gestational age, posterior placenta).
Fig. 1 shows a sketch of the measurement setup and coil positioning for phantom MRI [12] (Fig.1A) using the wearable 28-channel breast coil5 (“BraCoil”, Medical University of Vienna, Austria) (Fig.1B). A semi-flexible 18-channel coil (“Body18”, Siemens Healthineers, Erlangen, Germany) was used as a reference. The healthy pregnant volunteer was positioned feet first and laterally on the patient bed as instructed by medical personnel to avoid vena cava compression, with the wearable coil wrapped around the caudal part of the gravid abdomen (Fig.1C).
In fetal phantom MRI, 3D GRE scans were acquired to assess the coil coverage in comparison to the reference coil. In vivo, three-plane True Fast Imaging with Steady-State Free Precession (TRUFI) [13] data were acquired and a motion-correcting super-resolution algorithm [14] was applied to illustrate organ level details of the fetus. Fetal organs, and the amniotic fluid sac were manually segmented using MITK (2024) [15]. Example images showing fetal phantom GRE images in three spatial orientations in Fig.2 allow for a qualitative comparison of the coil coverage. As expected, the BraCoil’s coverage is limited in head direction but yields high superficial signal with an intensity gradient towards the center of the phantom. For in vivo imaging, therefore, the prescan normalization option was set to “broadband” (highest correction).
In vivo fetal TRUFI-MRI results in Fig.3 show that small structures in the liver, the lung and the femoral bone are clearly visible. In Fig.4, the isotropic motion-corrected super-resolution (0.89 mm) reconstruction results are presented which led to a detailed fetal model including brain (318 mm³), lung (85 mm³), liver (80 mm³), bladder (11 mm³), femoral bone and cartilage. Part of the fetal ankle bones, and upper limb humerus were also clearly reconstructed within this model. The amniotic fluid volume was measured to be 670 ml. The reduced FOV in BraCoil acquisitions can be advantageous for sequence planning as there are less fold-in artifacts from other body regions and saturation bands are no longer required. On the other hand, reduced FOV can be a drawback in patients presenting with a posterior placenta and advanced gestational age. Regions more distant from the coil suffer from lower signal as the combined use of the spine coil with customer coils was not allowed. With a dedicated fetal MRI coil either wrapping around the pregnant abdomen or used in combination with the spine coil, and an optimized (larger) coil element size, higher SNR and larger penetration depth can be expected. This would allow for a more holistic assessment of fetus and placenta simultaneously. MRI protocol optimization will be greatly facilitated by the developed fetal phantom. This work demonstrates the proof of concept of high-resolution fetal MRI in a comfortable lateral lying position using a wearable coil. In future work, a dedicated fetal coil will be developed based on learnings from more data acquired in pregnant women with varying gestational age.
Lena NOHAVA (Vienna, Austria), Rémi HATTAT, Juliette LEFEBVRE, Mbaimou Auxence NGREMMADJI, Marine BEAUMONT, Charline BERTHOLDT, Matthieu DAP, Olivier MOREL, Jacques FELBLINGER, Elmar LAISTLER, Bailiang CHEN
08:40 - 08:50
#46873 - PG017 Physically-Informed Deep Learning for Robust IVIM Quantification in Placental Diffusion MRI: Detecting Functional Alterations in Maternal Diabetes.
PG017 Physically-Informed Deep Learning for Robust IVIM Quantification in Placental Diffusion MRI: Detecting Functional Alterations in Maternal Diabetes.
Large-for-gestational-age (LGA) fetuses, commonly associated with maternal diabetes, are at increased risk for perinatal complications, including shoulder dystocia, neonatal hypoglycemia, and long-term metabolic dysfunction. Placental dysfunction is a key contributor to the pathophysiology of LGA and represents a valuable biomarker target for early risk stratification. Intravoxel incoherent motion (IVIM) analysis [1] of diffusion-weighted imaging (DWI) enables non-invasive estimation of microvascular perfusion and tissue diffusivity but suffers from limited reliability due to instability of the multi-parameter fitting process, particularly under clinically feasible acquisition schemes with sparse b-value sampling. This study investigates whether physically-primed deep neural networks that directly integrate b-value information into their architecture can better estimate placental IVIM parameters and improve sensitivity to disease-related changes in pregnancies affected by maternal diabetes.
Thirteen pregnant women were prospectively enrolled: five with uncontrolled maternal diabetes and eight healthy age-matched controls. MRI scans were performed on a 1.5T Philips Ingenia scanner (Philips Medical Systems, Best, The Netherlands) using a multi-channel abdominal coil. Placental diffusion-weighted images were acquired in the axial plane with a 2D spin-echo EPI sequence and fat suppression. Imaging parameters were as follows: repetition time (TR) = 4215 ms, echo time (TE) = 103 ms, slice thickness = 5.0 mm, inter-slice gap = 6.0 mm, in-plane resolution = 1.01 × 1.01 mm², and a 121 × 116 matrix. A total of nine b-values were used (0, 10, 20, 40, 80, 200, 400, 600, 1000 s/mm²), with a scan duration of ~531 seconds. No contrast agent was administered.
IVIM parameters—including D (tissue diffusivity), D* (pseudo-diffusion coefficient, associated with perfusion), and f (perfusion fraction)—were estimated for each subject using three methods: (1) classical segmented least-squares fitting (SLS-TRF), and two physically-informed AI approaches, (2) SUPER-IVIM-DC [2] and (3) SUPER-IVIM-DC-BOOT. Both AI methods leverage physically-informed supervised learning to stabilize IVIM parameter estimation from limited b-value data: SUPER-IVIM-DC enforces data consistency during training, while SUPER-IVIM-DC-BOOT extends this framework by explicitly incorporating b-values into the model architecture [3] and employing bootstrap resampling to enhance robustness. Parameter values were extracted from the placental regions of interest and averaged per subject. Welch’s t-test evaluated group differences for each method and parameter. The diabetic group was older on average than controls (35.5 ± 2.9 vs. 30.0 ± 5.5 years) and had slightly more advanced gestational ages at imaging (35.0 ± 2.8 vs. 30.3 ± 4.9 weeks). Classical IVIM analysis (SLS-TRF) did not identify significant differences between groups in any IVIM parameter.
In contrast, the SUPER-IVIM-DC model revealed a significant reduction in the D parameter among diabetic pregnancies (0.001827 ± 0.000086 mm²/s) compared to controls (0.001997 ± 0.000136 mm²/s; p = 0.03997), suggesting restricted tissue diffusivity in placentas affected by maternal diabetes.
The SUPER-IVIM-DC-BOOT model further demonstrated significant group differences in both D (0.001931 ± 0.000073 vs. 0.002043 ± 0.000104 mm²/s; p = 0.04365) and f (0.2896 ± 0.0428 vs. 0.3421 ± 0.0279; p = 0.0494), indicating impaired perfusion and diffusivity. D* did not differ significantly across methods. This study highlights the limitations of classical IVIM modeling in detecting placental dysfunction using limited DWI data. Physically-primed AI models, SUPER-IVIM-DC and SUPER-IVIM-DC-BOOT, provided enhanced parameter stability and sensitivity to pathological changes in maternal diabetes. The consistent detection of altered D and f parameters with AI methods suggests that diabetes-related microstructural and microvascular alterations can be non-invasively detected via improved IVIM quantification. The bootstrap-enhanced variant (SUPER-IVIM-DC-BOOT) further improved robustness and highlighted perfusion impairments not captured by traditional methods. Physically-informed AI models significantly outperform classical IVIM in detecting functional placental differences in LGA pregnancies associated with maternal diabetes. Their application enables more reliable extraction of diffusion and perfusion biomarkers from clinically practical DWI acquisitions. These methods hold promise for early, non-invasive identification of placental dysfunction in high-risk pregnancies and could facilitate personalized obstetric care.
Naama GAVRIELOV (Haifa, Israel), Moran GAWIE-ROTMAN, Abdel-Rauf ZEINA, Roni SHRETER, Esther MAOR-SAGIE, Rinat GABBAY-BENZIV, Moti FREIMAN
08:50 - 09:00
#47131 - PG018 Dynamic MRI assessment of functional and structural changes in the abdominal wall after hernia surgery.
PG018 Dynamic MRI assessment of functional and structural changes in the abdominal wall after hernia surgery.
Abdominal hernia is defined as a medical condition during which an internal organ or tissue protrudes through a weakness or defect in the abdominal wall muscles. This typically results in a visible bulge, which may increase in size when coughing or straining. As part of the surgical process, the surgeon pushes the herniated tissue back into its proper place and closes the gap in the abdominal wall through sutures. A synthetic mesh is generally placed over or under the weakened area to reinforce the abdominal wall and reduce the risk of recurrence. Despite advancements in surgical techniques, hernia repair surgery still faces significant challenges, particularly high recurrence rates reaching up to 70% [1]. This underscores the necessity for enhanced evaluation methods, especially for structural and functional outcomes. Dynamic magnetic resonance imaging (MRI) presents a non-invasive, non-irradiating tool that can be utilized to assess abdominal wall biomechanics both pre- and post-operatively.
Eleven patients scheduled for abdominal wall hernia repair volunteered to be included in the present study and to be MRI-scanned before and after surgery. Scans were acquired using a 3-Tesla MRI scanner (MAGNETOM Vida, Siemens Healthineers, Erlangen, Germany) while the patient was lying supine. Flexible RF coils were positioned on the top while bed-integrated coils were positioned on the bottom. Care was taken not to restrict abdominal movement. Static 3D MRI sequences were initially acquired so as to localize the ROI for the dynamic acquisitions. Axial and sagittal dynamic acquisitions (40 to 60s repetition) were then performed using a True FISP sequence over a 8 mm slice. Axial sequences were positioned pre-operatively at the largest hernia neck and post-operatively at matched locations. The sagittal plane was positioned at the midpoint of the hernia neck. Each subject completed three repeated tasks (deep breathing, coughing, and the Valsalva maneuver involving exhaling against closed mouth and nostrils for 8 seconds) following a brief training. Rectus abdominis and lateral muscles, linea alba, viscera area, and hernia sac were delineated as illustrated in Figure 1. The corresponding segmentations were used to compute abdominal displacement, strain, and shape variation. The analysis of axial images illustrated that coughing produced a mean hernia sac area increase of 128.4± 199.2%. Post-surgery, the distance between the rectus abdominis (i.e., where umbilical hernias are located preoperatively) was reduced by 13 mm (p ≤ 0.05) and muscle elongation was observed. Post-operatively, rectus abdominis thickness changes during breathing were inversely correlated with pre-operative hernia defect width (p ≤0.05).
The dynamic displacements of abdominal muscles are illustrated in Figure 2 for a pre-operative patient during the Valsalva maneuver. During the Valsalva exercise, the post-operative displacement of the lateral muscles was significantly larger in magnitude, indicating a greater inward movement, compared to the pre-operative displacement (p ≤ 0.05). For the rectus abdominis muscles, an almost significant increase in displacement was observed postoperatively during breathing (p = 0.09). Moreover, this displacement was negatively impacted by the size of the implanted mesh (p ≤0.05), indicating that larger mesh sizes were associated with reduced displacement of the rectus abdominis.
Valsalva exercise led to comparable area changes in axial and sagittal planes (35 and 33.6% respectively). Regarding the results in the sagittal plane, the largest post-operative change in displacement (in magnitude, either positive or negative) was observed near the surgical region of linea alba (p = 0.07). This investigation is the first to capture the real-time dynamics of the abdominal wall before and after hernia repair using dynamic MRI across two anatomical planes. Detailed in vivo visualization of the hernia sac, abdominal muscles, and viscera zone provided insights into post-operative functional restoration. While this approach enhances our understanding of biomechanical outcomes, the small sample size, patient variability, and absence of long-term follow-up limit the extrapolation of findings and clinical recommendations. Larger, longitudinal studies are needed to better define how these metrics could inform surgical strategies or predict recurrence. Dynamic MRI reveals detailed biomechanical changes in the abdominal wall following hernia repair. This imaging approach may support personalized monitoring and risk assessment.
Victoria JOPPIN (Marseille, Switzerland), David BENDAHAN, Catherine MASSON, Thierry BEGE
09:00 - 09:10
#47377 - PG019 Microstructural evaluation of rectal cancer surgical specimens using high-resolution advanced diffusion MRI (dMRI) and histological correlation.
PG019 Microstructural evaluation of rectal cancer surgical specimens using high-resolution advanced diffusion MRI (dMRI) and histological correlation.
T2-weighted imaging is the clinical standard for staging and restaging rectal cancer after neoadjuvant therapy (NAT), but has limited ability to distinguish tumour from fibrosis [1,2]. Diffusion MRI (dMRI), which reflects tissue microstructure and cellularity, offers greater potential for assessing treatment response [3–5]. This study is the first to look at the diffusion and T2 characteristics of different parts of the rectal wall using detailed quantitative MRI of whole total mesorectal excision (TME) specimens post-NAT. We evaluated whether diffusion and relaxometry MRI can distinguish tumour, fibrosis, and rectal wall layers - mucosa, submucosa, and muscle - with the aim of improving clinical decision-making.
All experiments were approved by the institutional ethics committee. TME specimens from four patients with rectal cancer were collected post-NAT. After 36 h in 10% buffered formalin and 4 h in 1xPBS, samples were mounted in cylindrical containers filled with Fomblin and scanned on a 9.4T Bruker Biospec system at 22 °C (86 mm transmit/receive). Samples were pseudonymized by the Champalimaud Foundation Biobank (Figure 1). We acquired 2D DTI with TR/TE = 11000/24 ms, 130 slices, 140×130 matrix, 0.5 mm³ isotropic resolution, 2 b0 images, b-values of 1500 and 3000 s/mm², and 15 directions. A 2D MSME sequence (TR = 25000 ms, 8 TEs from 10–80 ms) was acquired with matching geometry. All scans used fat suppression. Figure 2 shows representative ex vivo MRI data from a TME specimen, including T2-weighted contrast, averaged b = 3000 s/mm² signal, fractional anisotropy (FA), and mean diffusivity (MD) maps. The specimen was cut into 5mm sections (grossing stage); selected areas were paraffin-embedded and H&E-stained. Slides were digitized using a Philips Ultra-Fast Scanner 1.6 and matched to MRI slices using grossing images. We mapped the diffusion and kurtosis tensor parameters voxelwise using a linear least squares (LLS) algorithm in Matlab. We obtained maps of FA, MD, axial diffusivity (AD), radial diffusivity (RD), mean kurtosis (MK), axial kurtosis (AK), and radial kurtosis (RK). Subsequently, T2 maps were derived from MSME data by voxelwise mono-exponential fitting across eight echo times (10–80 ms) in Matlab. Finally, regions of interest (ROIs) for mucosa, submucosa, muscle layers, tumour and fibrous tissue were defined manually on MRI following MRI-histopathology correlation. A pathologist (23 years’ experience) identified tissues on histology, aligned to MRI using morphological landmarks, and confirmed by a radiologist (13 years’ experience). Figure 3 shows ROI placement guided by histology and dMRI (top), and spatial correspondence between MR contrast and tissue architecture. Tumour invasion into muscle (yellow/orange arrows) appears on FA maps as localized reductions in anisotropy (bottom). To evaluate differences between mean values across six tissue types of each diffusion parameter and T2 map, we used a Linear Mixed-effects Model on RStudio (v-2024.12.1), with FDR correction for multiple comparisons. Figure 4 demonstrates distinct diffusion and kurtosis profiles across rectal wall tissues, reflecting their unique microstructure. Muscle layers showed the highest FA, consistent with organized fiber orientation. Tumour and fibrous tissue also had elevated FA relative to mucosa. Tumour regions showed the lowest MD, indicating high cellularity, while fibrous tissue had higher MD. Kurtosis metrics (MK, AK) were highest in tumours, reflecting microstructural heterogeneity, and lower in fibrosis. T2 values showed limited contrast; mucosa and submucosa had slightly higher values, likely due to water content, but differences were minimal compared to diffusion metrics. High-resolution ex vivo dMRI enabled detailed characterization of rectal wall microstructure. Diffusion metrics clearly differentiated mucosa, submucosa, muscle, tumour, and fibrosis. FA maps highlighted muscle architecture, MD distinguished tumour from fibrosis, and kurtosis captured tumour heterogeneity. These results matched histology and prior ex vivo studies [5]. Diffusion imaging outperformed T2 mapping in tissue discrimination and detecting tumour infiltration, which showed limited contrast. While high-field fixed-tissue imaging may differ from clinical settings, the findings highlight the superior value of dMRI - particularly FA and MD - for identifying fiber disruption and microstructural changes associated with tumour invasion. Advanced diffusion MRI metrics (DTI and DKI) enhance tissue-specific characterization and more accurately detect tumour invasion into muscle layers, offering superior contrast and anatomical detail over T2 mapping for post-therapy assessment.
Ana FOUTO (Faro, Portugal), Mireia CASTILLO-MARTIN, Shermann MOREIRA, Noam SHEMESH, Laura FERNANDEZ, Hasti CALÁ, Nuno COUTO, Ignacio HERRANDO, Stephanie NOUGARET, Raluca POPITA, Jorge BRITO, Susana OURO, Miguel CHAMBEL, Nikos PAPANIKOLAU, Amjad PARVAIZ, Richard J. HEALD, Inês SANTIAGO, Andrada IANUS
09:10 - 09:20
#47815 - PG020 Bayesian multi-compartment analysis of transverse relaxation time and cellular proliferative activity in breast cancer on 3T.
PG020 Bayesian multi-compartment analysis of transverse relaxation time and cellular proliferative activity in breast cancer on 3T.
Precise estimation of cellular proliferative activity in breast cancer is central for monitoring the response to neoadjuvant therapy. The proliferative activity marker Ki-67 is widely used to inform prognosis, but relies on biopsy and provides limited spatial coverage [1, 2]. A non-invasive imaging biomarker that quantifies proliferative activity in the intra- and extra-cellular environments is hence highly desirable for treatment planning. Multi-compartment T₂ relaxation models are sensitive to compartment-specific microstructural properties of tissue, but are susceptible to noise [3, 4]. Recent Bayesian methods incorporate spatial priors across voxels to stabilise parameter estimation and enhance robustness under low SNR conditions [5]. We therefore hypothesise that intra- and extra-cellular T₂ from Bayesian model might provide a non-invasive, sensitive measure of proliferative activity, potentially offering an imaging biomarker for monitoring neoadjuvant therapy response.
We conducted a cross-sectional study on freshly excised tumour specimens from 20 patients (35 – 78 years) with invasive ductal carcinoma grades II (10) and III (10) (Figure 1). The study was approved by the North-West Greater Manchester East Research Ethics Committee (REC Reference: 16/NW/0221), with signed written informed consent obtained from participants prior to the study.
Data Acquisition: Quantitative transverse relaxation mapping of tumour specimens were acquired on a 3T MRI scanner (Achieva TX, Philips Healthcare) using a 32-channel receiver coil. Images were acquired using a multishot gradient and spin echo (GRASE) sequence [6], with 24 echoes, time of echo (TE) from 13 ms to 312 ms, echo spacing of 13 ms, repetition time (TR) of 9943 ms, field of view (FOV) of 141 × 141 mm2 and image resolution of 2.2 × 2.2 x 2.2 mm3. Tumour cellular proliferative activity was evaluated using the Ki-67 index, with more than 14% of tumour cell nuclei staining positive above the background considered as high Ki-67 [7] (9 high, 11 low).
Image Analysis: Voxel-wise overall T2time (T2, MONO) from single-compartment model was computed using a non-linear least squares method in MATLAB (R2023b, MathWorks Inc., Natick, USA). The intra- and extra-cellular T2 times (T2S and T2L) and volume ratio (f) were computed voxel-wise using the two-compartment model using a Bayesian algorithm [5]. Whole tumour delineation was performed on each specimen using MRIcron (v1.0.20190902, Colombia, USA) on conventional DWI images acquired at b = 800 s∙mm-2, with the necrotic regions excluded from the analysis. The parameters were computed as the mean within the whole tumour.
Statistical Analysis: Statistical analysis was performed in the SPSS software 27.0 (IBM Corp, Armonk, NY, USA). Shapiro-Wilk test for normality was performed on all relaxation parameters. Independent sample t-tests were conducted to compare the relaxation properties between high and low proliferating tumours. Correlation tests were performed on the T2, MONO, T2S, T2L and f against tumour diameter. A p-value < 0.05 was considered statistically significant. There was a significantly higher overall T2 time (p = 0.031) in high Ki-67 tumours (83.55 ± 7.38 ms) against low Ki-67 tumours (73.30 ± 11.30 ms) (Figure 2a, Table 1). There was a significantly higher intra-cellular T2 time (p = 0.047) in high Ki-67 tumours (73.52 ± 10.92 ms) against low Ki-67 tumours (61.30 ± 14.01 ms) (Figure 2b, Table 1). There was no significant difference in extra-cellular T2 time (p = 0.203) between high Ki-67 tumours (147.38 ± 8.84 ms) and low Ki-67 tumours (156.56 ± 19.16 ms) (Figure 2c, Table 1). There was no significant difference in volume ratio (p = 0.073) between high Ki-67 tumours (33.64 ± 8.33 %) and low Ki-67 tumours (41.65 ± 10.08 %) (Figure 2d, Table 1). There was a significant negative correlation in extra-cellular T2 time against tumour diameter (ρ = -0.50, p = 0.025, Figure 3a), however there were no significant correlations in the rest of the parameters against tumour diameter (Figure 3b-d, Table 1). The elevated overall transverse relaxation time in high proliferating tumours suggests reduced signal dissipation, potentially due to the diluted biochemical environment [8] from enhanced angiogenesis [9]. Rapid cell divisions in high proliferating tumours demand an up-regulated transport of amino acids for nuclear biosynthesis in the nucleus [10], leading to the dilution of the intra-cellular environment, resulting in increased intra-cellular transverse relaxation time [11]. The negative correlation between extra-cellular transverse relaxation time and tumour diameter might be due to the increased cellularity and reduced extra-cellular free water in larger tumours [12]. Bayesian-derived intra-cellular transverse relaxation time is associated with proliferative activities in breast tumours, potentially serving as a non-invasive imaging marker for neoadjuvant treatment monitoring.
Kangwa NKONDE (Newcastle, United Kingdom), Sai Man CHEUNG, Nicholas SENN, Jiabao HE
09:20 - 09:30
#47939 - PG021 Deregulation of lipid composition in the breast of BRCA1/2 genetic mutation carriers using chemical shift-encoded imaging.
PG021 Deregulation of lipid composition in the breast of BRCA1/2 genetic mutation carriers using chemical shift-encoded imaging.
Breast cancer is a major and expanding health challenge, despite significant improvement in survival rate [1]. Genetic mutation carriers of BRCA1/2 have over 30% increased risk of developing breast cancer and receive annual surveillance using DCE-MRI [2]. DCE-MRI is sensitive to tumour angiogenesis, however detects malignancies that are well under development. Deregulation of lipid composition, including monounsaturated, polyunsaturated and saturated fatty acids (MUFA, PUFA, SFA), has been shown in the breast of BRCA1/2 carriers using single voxel spectral edited MRS [3], and in the peri-tumoural region in patients [4]. Novel chemical shift-encoded imaging (CSEI) allows rapid lipid composition mapping of the whole breast, and the spatial distribution may further distinguish the disease state. We therefore hypothesise that lipid composition in the breast of BRCA1/2 carriers show deviation from healthy controls but no difference from patients with breast cancer, and determine the repeatability of CSEI.
Eighty-two premenopausal female participants, 30 BRCA1/2 carriers (age 45.1±8.3 years), 37 patients with invasive ductal carcinoma (age 45.6±7.0 years) and 15 age-matched healthy controls (42.7±7.2 years) participated in the study. Patients with a tumour size larger than 1 cm and have not had hormonal therapy or chemotherapy were eligible. Participants with diabetes or on long-term medications that might alter lipid composition were not eligible. The study was approved by the East of England – Essex Research Ethics Committee (Reference: 22/EE/0020), and written informed consents were obtained from all the participants (Figure 1).
Image Acquisition: All images were acquired on a 3 T whole-body clinical MRI scanner (Ingenia dStream, Philips Healthcare, Best, Netherlands). Lipid composition images were acquired from all participants using a 2D fast field echo sequence [5,6] with 16 echoes, initial echo time of 1.14 ms, echo spacing of 1.29 ms, repetition time of 60 ms, reconstruction voxel size of 2.0 × 2.0 mm2 and slice thickness of 3.0 mm, with subsequent repeated acquisition.
Image Analysis: Image analysis was conducted in MATLAB (R2020a, MathWorks Inc., Natick, MA, USA). The maps of the number of double bonds in triglycerides were computed from raw data, before subsequent quantification of MUFA, PUFA and SFA as a fraction of total lipids [5,6]. The delineation of tumour was conducted on the first echo of magnitude image, with reference to dynamic contrast enhanced images. The whole breast in BRCA1/2 and controls, and the whole breast and the peri-tumoural region in patients were the four regions-of-interest. The whole breast was defined to contain only adipose and fibroglandular tissue in BRCA1/2 and controls, and excluding the tumour in patients. The peri-tumoural region was defined as an annular ring of 16 mm (8 voxels) around the tumour. The median lipid composition from the regions-of-interest was subsequently computed.
Statistical Analysis: All statistical analysis was performed in the R software (v4.3.1, R Foundation for Statistical Computing, Vienna, Austria). Wilcoxon signed rank paired tests were performed for comparison of lipid composition in the whole breast and the peri-tumoural region in patients, with Wilcoxon rank sum tests performed between the whole breast of BRCA1/2, patients and controls. The within-subject coefficient of variation (%wCoV) was calculated as [standard deviation / mean] × 100%. A p-value <0.012 was considered to indicate a statistically significant difference for 4-group comparisons. There was a significantly higher MUFA (p=0.01) and lower SFA (p=0.01) in the whole breast of BRCA1/2 compared to controls. There was no significant difference in PUFA (p=0.03) between BRCA1/2 and controls (Figure 2, Table 1). There was no significant difference in MUFA, PUFA and SFA in the whole breast of BRCA1/2 against the whole breast nor the peri-tumoural region of patients (Figure 2, Table 1). There was a significantly lower MUFA and PUFA (both p<0.01), and higher SFA (p<0.01) in the peri-tumoural region compared to the whole breast in patients. (Figure 2, Table 1). The %wCoV in all four regions-of-interest were below 10.0% (Figure 3). Deregulation of lipid composition in the breast of BRCA1/2 carriers resembled the diseased group, serving as potential precursor of breast cancer. There was a decrease in MUFA and PUFA in the peri-tumoural region to support accelerated membrane synthesis for tumour growth [7], while an increase in SFA in the peri-tumoural region to avoid lipotoxicity and enhance chemoresistance [7]. CSEI has excellent repeatability in lipid composition Lipid composition in BRCA1/2 carriers showed similarity to patients. CSEI has excellent repeatability for accurate measurement of lipid composition in the breast.
Sai Man CHEUNG (Newcastle upon Tyne, United Kingdom), Kwok-Shing CHAN, Senthil RAGUPATHY, Zosia MIEDZYBRODZKA, Jiabao HE
|
Auditorium 900 |
|
"Friday 10 October"
Added to your list of favorites
Deleted from your list of favorites
B20
08:30 - 09:30
FT1-4 UNCONVENTIONAL SYSTEMS
FT1: Cycle of Technology
08:30 - 09:30
Low-field MRI.
Joseba ALONSO (Scientist) (Keynote Speaker, Valencia, Spain)
08:30 - 09:30
MR-LINAC.
Bas RAAYMAKERS (prof experimental clinical physics) (Keynote Speaker, Utrecht, The Netherlands)
08:30 - 09:30
MRI-PET.
Alessandra BERTOLDO (Director) (Keynote Speaker, Padova, Italy)
|
Espace Vieux-Port |
|
"Friday 10 October"
Added to your list of favorites
Deleted from your list of favorites
C20
08:30 - 09:30
FT3-3 QUALITY ASSESSMENT&CONTROL IN THE CLINICAL SETTING
FT3: Cycle of Quality
08:30 - 09:30
Beyond SNR - quantitative quality measures in advanced MR imaging.
Simone BUSONI (Senior Medical Physicist) (Keynote Speaker, Firenze, Italy)
08:30 - 09:30
Quality aspects in remote scanning.
Anton QUINSTEN (Keynote Speaker, Germany)
08:30 - 09:30
Workflow quality and operational efficiency in MR.
Susie HUANG (Keynote Speaker, Boston, USA)
|
Salle Major |
|
"Friday 10 October"
Added to your list of favorites
Deleted from your list of favorites
D20
08:30 - 09:30
GliMR
|
Salle 120 |
|
"Friday 10 October"
Added to your list of favorites
Deleted from your list of favorites
E20
08:30 - 09:30
MS6 - MRI of Hypothalamus in eating disorders
|
Salle 76 |
09:30 |
TIME FOR A BREAK
- Coffee and refreshments will be available at the cash bar.
|
09:50 |
"Friday 10 October"
Added to your list of favorites
Deleted from your list of favorites
A21
09:50 - 10:50
FT2 Plenary - Gained in translation
Adding value by crossing boundaries
FT2: Cycle of Translation
09:50 - 10:50
The translation from postmortem to in vivo.
Karla MILLER (Keynote Speaker, Oxford, United Kingdom)
09:50 - 10:50
Translational Cardiac Imaging at ultra-high field.
Laura SCHREIBER (Chair, Department Head) (Keynote Speaker, Würzburg, Germany)
|
Auditorium 900 |
10:50 |
TIME FOR A BREAK
- Coffee and refreshments will be available at the cash bar.
|
11:00 |
"Friday 10 October"
Added to your list of favorites
Deleted from your list of favorites
A22
11:00 - 12:30
FT2-2 - Microstructure MRI
FT2: Cycle of Translation
11:00 - 12:30
Inhomogeneous MT (ihMT) and myelin.
Olivier GIRARD (Ph.D.) (Keynote Speaker, Marseille, France)
11:00 - 12:30
Technical perspective on quantitative susceptibility mapping (QSM).
Sina STRAUB (Keynote Speaker, Bern, Switzerland)
11:00 - 12:30
White matter microstructure in the estimation of magnetic susceptibility.
Anders Dyhr SANDGAARD (Postdoc) (Keynote Speaker, Aarhus, Denmark)
|
Auditorium 900 |
|
"Friday 10 October"
Added to your list of favorites
Deleted from your list of favorites
B22
11:00 - 12:30
ET2-2 - Navigating Anesthesia
Challenges, Safety Strategies, and Non sedation Innovative Techniques
ET2: Cycle of Clinical Practice
11:00 - 12:30
Challenges and opportunities from the anesthesiologist's perspective.
Pierre SIMEONE (Keynote Speaker, Marseille, France)
11:00 - 12:30
Different methods/techniques used to MR scan patients and avoid anesthesia.
Darren HUDSON (Senior Lecturer) (Keynote Speaker, Exeter, United Kingdom)
11:00 - 12:30
MR Safety of anesthetized patients.
Roger LÜCHINGER (Keynote Speaker, Zurich, Switzerland)
|
Espace Vieux-Port |
|
"Friday 10 October"
Added to your list of favorites
Deleted from your list of favorites
C22
11:00 - 12:30
FT3 LT - Optimization of MR acquisition & processing
FT3: Cycle of Quality
11:00 - 11:02
#47566 - PG115 The Hidden Bias in Diffusion MRI: Effect of Inaccurate b-values from Imaging Gradients on Intravoxel Incoherent Motion.
PG115 The Hidden Bias in Diffusion MRI: Effect of Inaccurate b-values from Imaging Gradients on Intravoxel Incoherent Motion.
The effect of imaging gradients on b-values is an often-neglected bias in DWI. Imaging gradients can both increase and decrease the desired b-value [1], an effect which is not apparent in the metadata of DWI from clinical scanners. Typically, b-values are calculated using the classical formula b=γ^2 g^2 δ^2 (∆-δ/3). However, this formula only accounts for the diffusion weighting caused by diffusion encoding gradients and assumes zero contributions from imaging gradients. The actual diffusion weighting from a pulse sequence is calculated by including all of the gradients as follows [1]:
B=∫_0^TE q(t)⊗q(t)dt, (1)
where B is the b-tensor, and q(t)=γ∫g(t)dt, where g(t) is the effective gradient amplitudes as a function of time for the entire pulse sequence, which includes the inversion after the refocusing RF pulse. The b-value is given by the trace of the b-tensor, i.e. b = tr(B).
This discrepancy may be negligible in the context of other DWI applications, but has to our knowledge not been investigated for intravoxel incoherent motion (IVIM) analysis. IVIM typically employs b-values below 200 s/mm2, which may be more affected by imaging gradients [2]. The aim of this study is to investigate the effects of biased b-values on IVIM parameter estimates.
We investigated the propagated bias to IVIM parameters both in simulations and in vivo.
Two pulse sequences based on DWI sequences from two major MR vendors were simulated: one with large cross-term interactions between imaging and diffusion gradients, and one with minimal cross-terms (Figure 1). The actual b-values from an extensive set of 34 nominal b-values were calculated from simulations of various in-plane resolutions and slice thicknesses, and IVIM parameters estimated using a single-step NLLS fitting algorithm without noise with a range of different ground-truth values of the perfusion fraction f, pseudo-diffusivity D*, and diffusivity D.
A prostate dataset with 15 exams was used for in vivo evaluation. Data was acquired on a 3T Signa Architect (GE Healthcare, Milwaukee, WI, USA) with a Stejskal-Tanner pulsed gradient spin echo sequence. Reduced axial FOV with spatially selective excitation and CHESS fat saturation were used. Matrix size 160×80, in-plane resolution 1.5×1.5 mm2, slice thickness 3 mm, with TE 69.2 ms, and TR 5000 ms. Nominal b-values 50, 200, 800 s/mm2 were acquired in three directions with 5 repetitions each, including a nominal b0 with 15 repetitions. The actual b-values were calculated by recording the gradient waveforms for each shot of the pulse sequence. IVIM parameters were estimated using a typical segmented fitting approach with the b-value threshold set at 200 s/mm2 [3]. A notable variation in signal curves was observed between the two simulated pulse sequence designs (Figure 1). The sequence with large cross-terms showed a wider range of b-value variations across diffusion direction. This was also reflected in the estimated IVIM parameters (Figure 2). When using the nominal b-values, the bias in estimated f could be both positive and negative, while D* and D were consistently overestimated. Additionally, the bias decreased with the image resolution.
Similar inaccuracies were observed in the in vivo dataset. The actual b-values differed from the nominal by approximately 6% (Table 1). Figure 3 shows that f was underestimated by 0.7% (absolute percentage points), while D* and D were overestimated by 0.8 µm2/ms and 0.07 µm2/ms respectively. The simulations demonstrated that pulse sequence design plays an important role in the resulting actual b-values. This is problematic, since pulse sequence design may vary between commercial and research implementations of what can be considered a “standard” pulsed gradient spin echo sequence for DWI. This experimental variability underscores the need of using accurately determined b-values to ensure comparability of results across studies.
The in vivo analysis confirms that nominal b-values recorded in the DICOM metadata do not reflect the true diffusion weighting, as evident by the discrepancies between the nominal and actual b-values, and their impact on parameter estimates. In our dataset, actual b-values were generally higher than the nominal, whereas a previously published work demonstrated 6-15% lower b-values than expected [4], likely due to a different pulse sequence design. Neglecting imaging gradients in when calculating b-values leads to distorted and unpredictable systematic errors in IVIM results, and is not apparent to the users. The bias depends on pulse sequence design and imaging protocols, leading to substantial variations across studies and clinical implementations. Our findings reveal a need for data harmonization in future IVIM works to make results more comparable and reproducible.
Ivan A. RASHID (Lund, Sweden), Filip SZCZEPANKIEWICZ, Adalsteinn GUNNLAUGSSON, Lars E. OLSSON, Patrik BRYNOLFSSON
11:02 - 11:04
#47902 - PG116 Faster multi-parameter mapping via combined sensitivity estimation and inter-scan motion correction.
PG116 Faster multi-parameter mapping via combined sensitivity estimation and inter-scan motion correction.
Inter-scan motion causes error in quantitative MRI (qMRI) if the receive coil sensitivity modulation is assumed consistent across volumes. Rapid, low resolution, calibration data (‘s-maps’) acquired prior to each volume can mitigate these [1] by removing relative sensitivity modulation across positions. qMRI protocols typically use parallel imaging to ensure feasible scan times, high spatial resolution and whole brain coverage. Here, using a common qMRI protocol, Multi-Parameter Mapping (MPM), we use s-maps to both unfold and correct inter-scan motion artefacts in magnetisation transfer saturation (MTsat) maps [2], which rely on the combination of three distinct volumes. We further demonstrate that care must be taken to ensure data are handled consistently, e.g. when pre-whitening or combining images across coils.
An MPM protocol (Fig.1) was acquired at 7T 3 times, twice in position 1 (scan/rescan) with no intentional motion, and a third time in position 2 following large, intentional motion. Combining data within positions provides a reference against which to assess the effect of inter-scan motion introduced when data are combined across positions.
Conventionally, integrated reference data, i.e. a central fully sampled portion of k-space within the accelerated volumes, are used to unfold the accelerated MPM volumes. MORSE-CODE [3], a fast and robust regularised SENSE [4] image reconstruction formalism, used coil sensitivities estimated either from these integrated data or the s-map data.
Signal-to-noise can be improved by using noise calibration data to estimate the coil covariance and whiten the data prior to unfolding. Noise estimates are acquired as part of each acquisition, i.e. independently for the s-map and under-sampled data. We investigated the impact on image quality of using acquisition-specific (inconsistent) or consistent noise estimates.
We also investigated the impact of combining the individual channel data of the s-map acquisition as root-sum-of-square (RSOS, default on the scanner host) or via sensitivity-weighted combination consistent with the MORSE-CODE unfolding scheme.
Quantitative parameter maps were estimated using the h-MRI toolbox [5]. The scan-rescan estimates from position 1 were used to assess the image reconstruction performance. A coefficient of variance (CoV) measure was computed as the difference between the scan/rescan MTsat estimates with respect to their mean at each voxel within a grey and white matter mask.
Data were combined across positions (PD- and T1-weighted from position 1 with MT-weighted from position 2) to manifest inter-scan motion artefacts in an MTsat map. MTsat maps were estimated with no inter-scan motion correction, and again with inter-scan motion correction using either the RSOS or sensitivity-weighted MTsat maps. CoV was again computed, now as the difference between these maps (with/without motion correction) and a reference MTsat map computed at position 2. The participant moved approximately 20 degrees about the z-axis between positions.
Whitening using acquisition-specific noise estimates led to visible aliasing artefact after unfolding (Fig. 2). When whitened consistently, the integrated reference or s-map sensitivities unfolded images with equivalent quality. Any residual artefacts did not propagate to the estimated parameter maps (c.f. MTsat map, Fig. 2). CoV estimates showed scan-rescan reproducibility was equivalent (Fig. 3) as long as consistently whitened.
As expected, inter-scan motion led to substantial artefact in the MTsat map (Fig. 4), and was reduced by the established correction scheme. Neither coil combination approach fully removed the artefact but CoV analysis showed that the sensitivity-weighted coil combination agreed best with the reference MTsat map (Fig. 4). qMRI protocols often rely on calibration data. Here we show that calibration data used for inter-scan motion correction can additionally be used to estimate sensitivities needed to unfold accelerated datasets. Protocols that utilise multi-echo readouts and estimate sensitivities from integrated reference data will particularly gain from this approach since the single echo nature of the ‘s-map’ data enables a much shorter TR than integrated reference data.
However, care must be taken to whiten the data consistently. Temporal, heating or motion effects modestly influence the noise covariance but the resulting whitening matrix can substantially alter the sensitivities leading to residual artefacts if inconsistent with the data they are used to unfold.
Using a consistent coil-combination approach for the calibration and target data also improves the quality of the inter-scan motion correction scheme. However, residual effects such as position-specific transmit fields remain uncorrected. qMRI protocols can be made more efficient, without degrading reproducibility, by replacing integrated reference data with ‘s-map’ data used to correct inter-scan motion.
Benjamin JAMES (London, United Kingdom), Barbara DYMERSKA, Martina CALLAGHAN
11:04 - 11:06
#47049 - PG117 Susceptibility-weighted MRI with optimized phase mask for central vein sign detection in the spinal cord at 7T.
PG117 Susceptibility-weighted MRI with optimized phase mask for central vein sign detection in the spinal cord at 7T.
Susceptibility-weighted imaging (SWI) has shown great potential in the brain to identify the central vein sign (CVS), which refers to the presence of a vein within multiple sclerosis (MS) lesion and was shown to be a specific marker of this pathology [1]. This biomarker was recently added to the clinical recommendations for MS diagnosis for brain MRI [2]. Its role and presence in the spinal cord was reported in ex vivo studies [3] but was, however, poorly determined in the spinal cord where visualization is made difficult by the small size of the veins [4]. Indeed, anisotropic resolutions typically used in the cord lead to large partial volume effects, as the volume of the vein only represents less than 20% of the total voxel volume, whereas it can reach 100% in the brain (Table 1).
In order to remove this obstacle, this study introduces an optimization of the SWI phase mask to isolate contributions linked to the presence of veins with orientations and sizes [1] representative of those expected in MS patients with spinal cord lesions. The proposed SWI post-processing was applied to detect CVS in the spinal cord.
*MRI data:
A retrospective analysis was performed on multi-slice multi-angle axial 2D T2* multi-echo gradient-echo [5] data, acquired at the cervical level on a 7T Magnetom (Siemens Healthcare, Erlangen, Germany) on 13 MS patients, with measured angles between the slice and z-axis between -36° to 18°, and sequence parameters: FA = 50°, 2 averages and two different spatial resolutions:
- 0.18 x 0.18 x 2 mm3: TE = 5.2, 9.43, 13.66, 17.89 ms / TR = 500, TA = 10 min 26 s
- 0.27 x 0.27 x 2.5 mm3: TE = 5.02, 9.01, 13 ms / TR = 565, TA = 6 min 36 s
*Theory of phase calculation in the presence of a vein:
The intra- (ΔφIV) and extravascular (ΔφEV) phase contributions of a voxel containing a vein [4] were calculated according to:
ΔφIV = -2π γ Δχ B0 . (cos²θ - 1/3).TE , and
ΔφEV = -2π γ Δχ B0 . sin²θ . cos2ψ . (R0⁄r)² . TE
with Δχ: difference in susceptibility between the deoxygenated blood and the lesion; θ: angle between the vein orientation (of radius R0) and B0; ψ: angle between B0 and the projection of a vector indicating the position of a point in the extravascular space in a plane perpendicular to the vein; r: distance between this same point and the vein center.
*Phase mask optimization for CVS identification:
1) Calculation of ΔφIV and ΔφEV for TE, voxel dimensions and orientations corresponding to in vivo GRE acquisitions (0 to 15° relative to the B0-field, which are the most common orientations), as well as: vein diameter = 0.3 à 0.5 mm; variation of vein position within the voxel; θ = 45° to 135° to enhance vessels visible over multiple voxels.
2) Post-processing of the SWI to attenuate the signal from voxels with these phases in the spinal cord, using the CLEAR-SWI toolbox [6].
3) Inspection of lesions identified by neuroradiologists [7] after application of the phase filter and counting of lesions with CVS. Figure 1 shows histograms of the calculated phases of voxels containing a vein with all possible configurations of parameters described in Methods. Results reveal how vein positions and voxel dimensions have a substantial impact on phase signal. For (A) and (B), all phases are within the range [-0.55:0.1] and [-0.25:0.05] radians, respectively, allowing to use band-pass filtering of the phase in the SWI post-processing.
Figure 2 shows the post-processing steps of the proposed SWI method, displaying the SWI as well as the phase mask at different steps. By applying phase masks only to spinal cord voxels with the calculated phases, and in groups of 4 or more adjacent voxels, indications of the presence of medullary veins were observed. The blue arrow shows an indication of CVS in a spinal cord lesion which was not observed in the T2* GRE and was surrounded by noisy signal with ‘brain-like’ processing.
Figure 3 shows examples of healthy control and a representative MS patients T2* GRE and after using the proposed optimized SWI processing. A strong attenuation is observed due to voxels containing veins, with a distribution similar to expected vasculature [8]. In particular, radial veins and a venous ring at the surface of the cord can be observed in some slices. Of the 75 lesions identified by experienced neuroradiologists, 43% showed indications of CVS (blue arrow in Figure 3), which is close to the proportion observed in the brain [1]. The observation of CVS in the spinal cord opens up diagnostic and prognostic perspectives. The small veins present in the spinal cord require different processing than in the brain to isolate their phase contributions without enhancing noise, which would be the case with typical SWI processing. Further work will include a larger cohort and reproducibility evaluations. Similar optimizations may be required for other resolutions and applications (such as brain), in order to isolate voxels of interest in the presence of veins that are smaller than the voxel size.
Aurelien DESTRUEL (Marseille), Sarah DEMORTIERE, Maxime GUYE, Jean PELLETIER, Virginie CALLOT
11:06 - 11:08
#46387 - PG118 FLAIR with robust CSF suppression by optimal control.
PG118 FLAIR with robust CSF suppression by optimal control.
Fluid-attenuated inversion recovery (FLAIR) [1-4] is an essential component of routine brain MRI protocols [5], characterized by the suppression of cerebrospinal fluid (CSF) using an inversion pulse. However, when the inversion is imperfect, residual bright signal from poorly inverted magnetization can appear in the final images, potentially mimicking or obscuring pathology. Artifacts are particularly common in regions affected by strong inhomogeneities in either the RF or B0 field. RF inhomogeneities often occur at the edges of the field of view, while B0-inhomogeneities are prominent near air-tissue and bone-tissue interfaces. Both effects are more severe at higher field strengths. Adiabatic pulses [6], routinely used in clinical FLAIR protocols for magnetization inversion, provide robustness against RF field inhomogeneities by design, once the RF amplitude exceeds the adiabatic threshold. Inversion capability in the presence of off-resonance is characterized by the bandwidth of the pulse. However, the drawback is a usually prolonged pulse duration. The aim of our study was to improve the robustness of FLAIR against both RF and B0 inhomogeneities by applying robust inversion pulses designed by time-optimal control.
Pulse Optimization: The FLAIR inversion pulse was optimized for time efficiency and robustness to B0 and B1 inhomogeneities using ensemble-based time-optimal control [6]. The constraints included:
- Non-selective inversion targeting Md=(0,0,-1).
- B0 robustness for ±2.4 ppm at 3T.
- B1 robustness for 80% to 115% of nominal RF amplitude, with a max RF amplitude of 20 μT (handled as box constraint).
- Time discretization of 0.01 ms.
- Initial random amplitude and phase over 10 ms.
The Bloch equations were solved using symmetric operator splitting [7], and gradient calculations employed adjoint calculus [8]. The final optimized RF pulse had a duration of 2.01 ms and was integrated into the FLAIR sequence without altering other parameters.
Data Acquisition: Scans were conducted on a 3T Philips Elition MRI with a 32-channel SENSE head coil. The study was approved by the Clinical Research Ethics Board, and informed consent was obtained from all participants.
Phantom Scans: A cylindrical phantom (13 cm diameter, 17 cm length) with a smaller cylinder (1.5 cm diameter, 9.5 cm length) filled with 0.05 mmol/ml gadolinium solution was used. The smaller cylinder was oriented perpendicular to B0 to induce field inhomogeneities.
In Vivo Scans: Standard FLAIR and robust FLAIR scans were compared in three healthy participants. Imaging parameters were sagittal 3D with TI/TR = 2400/8000 ms, voxel size 1.2 × 1.2 × 1.2 mm³, reconstructed to 0.67 × 0.67 × 0.67 mm³, and a field of view of 256 × 256 × 170 mm³. The only difference between the scans was the inversion pulse: a 17.16 ms hyperbolic secant for the conventional FLAIR, as used within the clinical protocol, and the optimal control pulse with a duration of 2.01 ms for robust FLAIR. CSF suppression was assessed visually. Fig. 1 illustrates the inversion efficiency across a range of B0 inhomogeneities and RF amplitudes. The adiabatic pulse maintains an efficiency of approximately 0.9 for B0 inhomogeneities between -2.4 and +2.4 ppm, but its efficiency drops rapidly outside this range. In contrast, the robust pulse achieves nearly perfect inversion (close to 1) over a broader range of B0. Additionally, it is less sensitive to reduced RF amplitudes, maintaining nearly perfect inversion even at 70% of the nominal RF amplitude.
In phantom scans, using the adiabatic pulse, regions of strong B0 inhomogeneities show inadequate inversion, leading to bright signal artifacts (Fig. 2, top). In contrast, FLAIR with the robust pulse show perfect inversion throughout the entire phantom, with dark areas near the Gd-filled cylinder due to very short T2 relaxation times in those regions (Fig. 2, bottom).
In human volunteers, the standard adiabatic inversion pulse leads to bright signal artifacts at the base of the frontal lobes, a region known for strong B0 inhomogeneities (Fig. 3). The robust pulse significantly reduces these artifacts compared to the conventional scan. FLAIR is critical for detecting brain pathologies such as tumors, infections, trauma, and MS, but remains prone to artifacts near the skull base and from implants. These distortions often obscure clinical findings. While parallel transmission reduces RF inhomogeneities, B0-related artifacts persist despite advanced shimming techniques. The proposed optimal control inversion pulse addresses this by offering robustness to both B0 and B1 inhomogeneities, without requiring patient-specific mapping or additional scan time. The optimized pulse eliminates artifacts without changes to hardware or acquisition protocols, enabling seamless clinical integration. Though demonstrated at 3T, the design can be readily adapted to other field strengths.
Christina GRAF (Vancouver, Canada), Alexander JAFFRAY, Armin RUND, Stefan STEINERBERGER, David LI, Alexander RAUSCHER
11:08 - 11:10
#46708 - PG119 Simulation of dynamic B0 with Phase Distribution Graphs.
PG119 Simulation of dynamic B0 with Phase Distribution Graphs.
MRI simulations are valuable tools for developing new sequences, understanding imaging mechanisms, optimizing parameters, generating machine learning training data and more. These simulations typically assume constant physical properties of the simulated tissues. This does not fully describe clinical measurements where these properties can change, especially due to movement. One of the most noticeable effects is a varying magnetic field, influenced by the breathing of the subject. The resulting fluctuations can be measured even in neuroradiological imaging. In this work we derive the necessary theory and implement a simulation of dynamic B0, with the goal of moving even closer to in-vivo measurements.
With this work we strive to extend our simulation based on Phase Distribution Graphs[1], which currently is the fastest existing simulation for physically accurate simulation of imaging sequences. It is part of MR-zero[2] and recently has gained the capability of simulating motion[3]. Due to changes in the spatial distribution of susceptibility, motion can influence the homogeneity of the magnetic fields, especially the main magnetic field B0. This can in turn affect measurements even in the brain and introduce artifacts in long measurement series.
To accelerate the simulation of dynamically changing B0, we analytically derived the accumulated phase that results from B0 inhomogeneities. This is done by defining B0 at fixed time points as input maps and applying closed-form equations for the resulting phase, which in turn is included in the signal equation. Further integration into the simulation also ensures correct interaction with RF pulses for a complete physical model of the magnetization dynamics.
The influence of dynamic B0 is examined with the simulation of a balanced SSFP and a FLASH sequence. First, 120 B0 maps during multiple breathing cycles were measured using the DREAM[4] sequence. They were then combined with a quantified[5] phantom to complete the simulation data. With this data, two simulations were computed: 1. A bSSFP with 50° pulses, 10 ms repetition time, alternating RF pulse phases and an α/2 preparation pulse. 2. A 7° FLASH sequence with a repetition time of 20 ms and quadratic RF phase increment of 84°. Both sequences have centric phase encoding reordering and a resolution of 64×64, while the simulation internally runs at a higher resolution of 96×96. The resulting signals were reconstructed into images using a simple Fourier transform.
Both sequences were simulated twice, once with the dynamically changing B0 and once with the average, static B0 that was acquired during quantification. To amplify the effects of dynamic B0 for visualization purposes, the breathing cycle was accelerated such that its first half of fully breathing in was completed in the time of the acquisition of each sequence. The overall B0 map is shown in Figure 1. It is relatively with a standard deviation of 15 Hz. Compared to it, the fluctuations in the dynamic B0 maps are low. In figure 2, they are shown relative to the average B0. They deviate from it in the range of ±6 Hz. The deviation is mostly but not fully homogeneous, meaning that not only the amplitude but also the shape of B0 changes throughout a breathing cycle.
Comparing the simulation of the FLASH sequence between static and dynamic B0 shows a different phase, which is due to the different B0 inhomogeneities during acquisition. Figure 3 shows no higher order effects, which can be a result of the interaction between accumulated phase and RF pulse, are not visible. FLASH sequences measure mostly newly excited magnetization, which has a trivial dependency on B0 inhomogeneities.
In Figure 4, a different picture emerges. The simulated bSSFP sequences produces an image which is the combination of many different magnetization pathways. As an RF pulse can excite and refocus at the same time, these pathways all accumulate the phase of B0 inhomogeneities in different ways. In combination with dynamically changing B0, this leads to more complex artifacts. The result are different ringing patterns in the reconstructed image. In addition, the phase of bSSFP images is expected to be close to zero through the interference of excited and refocused magnetization pathways. With dynamic B0, refocusing must not completely revert the phase of magnetization and the deviations can be larger. We introduced analytical dynamic B0 to our state-of-the-art MRI simulation. We demonstrated the influence of B0 on FLASH and bSSFP brain measurements. Although small, noticeable changes to the resulting contrasts could be detected. Measurements that rely on spin echoes and similar mechanisms will show more complex dependencies on the varying B0 inhomogeneities than others. With the introduction of dynamic B0, the accuracy of the simulation could be improved substantially. This can be used for better understanding of artifacts in time-series acquisitions or long measurements.
Jonathan ENDRES (Erlangen, Germany), Simon WEINMÜLLER, Moritz ZAISS
11:10 - 11:12
#47703 - PG120 3D printing of subject-specific passive shims to improve MRI for in vivo subjects.
PG120 3D printing of subject-specific passive shims to improve MRI for in vivo subjects.
Magnetic resonance imaging (MRI) relies on a homogeneous magnetic field (B0) for optimal image quality. Although MRI scanners are designed for inherent field uniformity, patient-specific anatomical variations and medical implants introduce local B0 inhomogeneities, which degrade imaging performance [1-4]. To counteract these inhomogeneities, B0 can be adjusted, or "shimmed", for greater uniformity.
Active shimming involves applying electric current through dedicated coils to adjust the B0 field, typically addressing up to the second order of field inhomogeneities. However, subject-specific anatomies introduce complex distortions that challenge conventional active shimming. Moreover, higher order shim solutions can interfere with MRI equipment and require custom hardware [5-8].
Passive shimming strategically places magnetizable materials (e.g., ferromagnetic, diamagnetic) to enhance field uniformity [9]. However, the use of passive shimming to correct subject-induced B0 variations has not been widely adopted due to its labor intensity, high costs, and challenges associated with higher order field distortions [10-12].
This research introduces an innovative 3D printing technology to produce customized passive shims aimed at correcting field inhomogeneities induced in vivo by specimens. Unlike conventional methods, our powder-binder jetting technology enables the efficient production of shims with complex geometries without additional costs or extended production times by allowing precise deposition of ferromagnetic material, generating higher order spherical harmonic terms to optimize B0 homogeneity (Fig. 1) [13].
Simulations and Scan:
Field maps of female Fischer rats were acquired on a 9.4T animal MRI scanner (Fig. 2A, B). These maps served as input for a custom-developed shimming algorithm (MATLAB) designed to minimize the standard deviation of B0 by employing the MEIGO global optimization toolbox [14]. Fmincon functioned as the local solver to tackle the nonlinear optimization problem:
Std= √(1/(N-1) ∑|(A*ink+unshimmed_vec )-µ|² ) (Eq. 1)
with
µ= 1/N ∑(A*ink+unshimmed_vec) (Eq. 2),
where unshimmed_vec is the unshimmed field map converted to a vector, A is the sensitivity matrix describing the influence of each voxel, N is the number of voxels and ink is the concentration of ferromagnetic ink. The concentration in each voxel to achieve optimal homogeneity is calculated and converted into a grayscale printable CAD design.
3D printing:
The design was fabricated via a modified powder-binder 3D printer. The primary build material was polymethyl methacrylate powder. Thermal printheads were employed for the ferromagnetic ink (containing magnetite nanoparticles at 30 wt%) and the binder ink (2:1 volume mixture of acetophenone and butanone). The 3D-printed shim was scanned via a Phoenix Nanotom (CT). The reconstruction was performed with Phoenix datos|x with subsequent image processing in Avizo 2019.1 (Fig. 2C). Simulations and Scan:
Simulations were performed for the brain as a region of interest. The standard deviations of both unshimmed and shimmed fields were compared. The results of the simulation demonstrated a potential improvement of 29% in B0 field homogeneity (Fig. 3A). After printing and measuring the field maps with and without shim, an actual improvement of 21% in standard deviation was observed (Fig. 3B). This difference may arise from several factors:
1. Positioning variability: Misalignments between the shim and the rat during MRI scanning might reduce shimming efficacy. This could potentially affect the correction for higher order inhomogeneities, but can be avoided in future measurements by 3D printing a holder for the shim.
2. Printing imperfections: Slight deviations in ferromagnetic ink deposition (e.g., droplet spread or inhomogeneous nanoparticle distribution) could affect the shim’s magnetic properties. This deviation is an inherent characteristic of the 3D printer and cannot be accounted for in the simulations.
3. Scanning: Noise in MRI field maps may introduce small variations in quantifying the field difference. This can be minimized by increasing the resolution or the number of scan averages.
3D printing:
It is essential to accurately control the amount and positioning of printed ferromagnetic ink. The ferromagnetic ink distribution within printed shims was visualized for voxels of 1 mm³ to verify this precision. The average measured dimensions in the x-direction were 0.97 mm ± 0.08 mm and 0.98 mm ± 0.08 mm in the y-direction. The analysis confirmed dimensional inaccuracies below a 10 µm threshold, with the resolution set to 300 µm in the x/y-directions and 100 µm in the z-direction (print layer height) [13]. Our results prove that 3D printed passive shims can effectively homogenize the magnetic field in a scanner for in vivo species. Future research will focus on testing the technique on larger specimens (e.g., monkeys, dogs) with complex anatomical structures to further evaluate its efficacy.
An VANDUFFEL (Leuven, Belgium), Hanne VANDUFFEL, Cesar PARRA CABRERA, Błażejczyk KASIA, Shannon HELSPER, Quentin GOUDARD, Uwe HIMMELREICH, Dimitrios SAKELLARIOU, Rob AMELOOT
11:12 - 11:14
#47363 - PG121 Relaxivity of gadolinium-based contrast agents in cerebrospinal fluid at 3T.
PG121 Relaxivity of gadolinium-based contrast agents in cerebrospinal fluid at 3T.
Interest in cerebrospinal fluid (CSF)-flow and distribution has grown with the proposed glymphatic system, a potential brain waste clearance system [1]. In this context, T1-mapping before and after intrathecal injection of a gadolinium-based contrast agent (GBCA) has been used to study the flow of CSF [2]. Repeated T1-mapping enables quantification of gadolinium (Gd) concentrations in CSF and brain tissue [2], allowing modeling of the glymphatic flow. However, accurate quantification of the Gd concentration depends on the relaxivity of the GBCA. The relaxivity of a GBCA is dependent on temperature, magnetic field strength and medium it is measured in, but has never been estimated in CSF.
The aim of this study was to estimate the T1-relaxivity of two GBCAs in CSF at 3T. For comparability with previous studies, due to their similar properties, we also estimated the T1-relaxivity in an isotonic solution.
Using a phantom model (figure 1), we calculated the relaxivity of the two GBCAs gadobutrol (GADOVIST® Bayer Pharma) and gadoteric acid (DOTAREM®, Guerbet) in both CSF and an isotonic solution (Ringer-Acetat Baxter Viaflo, Baxter). CSF was acquired from patients investigated for idiopathic normal pressure hydrocephalus. CSF-samples were discarded if albumin, erythrocytes or cell counts were outside the reference value. The CSF used in the phantom experiment were a mix of equal portions from different subjects. CSF and isotonic solution were diluted with either gadobutrol or gadoteric acid to eight Gd concentrations between 0-1mM. For comparability, both GBCAs in both mediums were scanned simultaneously. To account for T1 variability with temperature, the phantom was insulated in Styrofoam and heated to 37.5 °C before the scan, with temperature measured during the scan.
Phantom measurements were done on a 3T MRI (Signa Premier; GE Healthcare) with a 48-channel head coil. T1-mapping was performed using the variable flip angle method [3], acquired with a 3D fast spoiled gradient echo sequence with isotropic acquisition of 1 mm, 100 slices, field of view 256x256, TR=7.2, TE 2.9, flip angles α1=2°, α2=12°, α3=7°, α4=16°, α5=20°. B1-mapping was performed using the Bloch-Siegert shift approach [4].
T1-maps were generated per voxel using a nonlinear least squares approach optimized using the Levenberg-Marquardt method. The relaxivity was estimated from the slope of the linear regression between the relaxation rate (1/T1) and the Gd concentration, with errors presented as the standard error of the coefficient. All relaxivity values are given in unit of L mmol-1s-1. Differences in relaxivity between the CSF and isotonic solution was tested with a regression model, including interaction terms for the slope and intercept between CSF and the isotonic solution. A p-value of < 0.05 was considered statistically significant. The relaxation times for the 0 mM Gd samples were 4506 ms for CSF and 4385 ms for the isotonic solution. The linear regressions used for the relaxivity estimations are presented in figure 2. The relaxivity of gadobutrol was 3.98 ± 0.16 in CSF and 2.95 ± 0.12 in the isotonic solution (p<0.001). For gadoteric acid the corresponding values were 2.88 ± 0.11 in CSF and 2.78 ± 0.08 in the isotonic solution (p=0.49). The mean temperature during the scan was 37.4 (range 37.1- 37.6) °C. We present the T1-relaxivity in CSF and an isotonic solution for two GBCAs in a physiological temperature at 3T. Our results show that the relaxivity in CSF differ from an isotonic solution for gadobutrol but not for gadoteric acid. This difference is of importance to note for future research on the glymphatic system utilizing T1-mapping.
No previous study has measured the T1 relaxation time of CSF in vitro. The T1 relaxation time in water has previously been measured to 4420 ± 103 ms [5], closely resembling our measure of 4385ms in the isotonic solution. The relaxivity in water has previously been estimated to 3.2 ± 0.3 and 3.3 ± 0.2 for gadobutrol [5, 6] and 2.8 ± 0.2 for gadoteric acid [6], both being near our relaxivity estimates of 2.95 ± 0.12 and 2.78 ± 0.08, for gadobutrol and gadoteric acid respectively in the isotonic solution. Our isotonic solution results thus resemble those of previous studies, speaking towards the validity of our measurements. We estimated the T1-relaxivity in CSF at 3T for gadobutrol and gadoteric acid. There was a difference in the relaxivity between CSF and water for gadobutrol, but not for gadoteric acid. In future studies on CSF-flow using T1-mapping post contrast injection, we recommend using a relaxivity value specific for CSF.
Sofia BEHNDIG (Umeå, Sweden), Anders GARPEBRING, Daniel DAHLGREN LINDSTRÖM, Jan MALM, Anders WÅHLIN, Anders EKLUND
11:14 - 11:16
#47632 - PG122 Proof of concept study of UTE-based dosimetry maps in head and neck cancers.
PG122 Proof of concept study of UTE-based dosimetry maps in head and neck cancers.
The use of MRI in radiotherapy planning has increased over the years as MRI represents a non-ionizing modality providing both an improved contrast between soft tissues compared to the classically used CT scan and an opportunity for personalized therapy. However, a limitation toward MRI-only dosimetry is the absence of direct physical link regarding the electron density (ED) of the tissues that is provided by the CT scan. This information is required by dosimetry algorithms to compute the dose distribution. Efforts have been made to overcome this limitation with a focus of the community oriented toward the generation of synthetic CT images using artificial intelligence algorithms [1]. Another approach is to rely on quantitative information acquired through MRI to approximate the tissues ED. Demol et al. [2] showed that dosimetry based only on the hydrogen content of the tissues performed as well as ones based on CT scanner. Seco et al. [3] provides an equation for the ED as a function of the atomic composition of the tissues.
MRI sequences such as the Ultrashort Echo Time (UTE) sequence can be used to obtain a signal related to the proton density contained within the tissues. Thus, it would be possible to derive ED maps from UTE images with a tissue correction provided by the mass density that would account for the bone that is difficult to measure in MRI.
A phantom with variable proton density was designed using water tubes diluted with D2O in various proportion. UTE images were acquired on a 3T Siemens VIDA (Siemens Healthineers, Erlangen, Germany). The average signal value in each tube was linked to the corresponding theoretical proton density using linear regression.
A study was designed using 3T MR and CT images of thirteen patients with various tumors locations. The MRI protocol included a Spiral VIBE UTE sequence which was set up to recover a signal proportional to the proton density of the tissues (flip angle = 5° and TE = 0.03ms). Preprocessing included N4 bias field correction of the MR images and registration of CT in the MRI space using the SimpleITK [4].
The UTE signal of the tissues was expressed relatively to the UTE signal of pure water. The ED value was obtained by using the following equation based on the results of Demol et al. [2] and Seco et al. [3]: ρ_(e_tissue) = (ρ_(H_tissue) + ρ_tissue)/2 with ρ_(e_tissue) the ED of the tissue relative to water, ρ_(H_med) the UTE signal relative to water and ρ_tissue the tissue's mass density.
A categorical mask of the main tissue types (air, bone, fat and soft tissues) was derived from the CT through Hounsfield Unit thresholding. The mask was used to apply the tissue correction based on mass density values from the literature [3]. Fig 1 shows the final dosimetry maps: UTEtissues refers to the maps with tissue correction while UTEwater refers to maps where water mass density was used in the whole body. Values represent ED multiplied by 100 to respect the DICOM formalism.
Dosimetry plans used for the treatment of each patient were recomputed without re-optimization using MONACO (Elekta AB). Resulting dose maps were analyzed through dose differences at 95% of tumoral volumes (TVs) and at 2% organs at risks volumes (OaRs). Gamma pass rate at 3%/3mm and 2%/2mm were computed using PyMedPhys[5] Measurements of the UTE signal were plotted against the theoretical proton density within the tubes. Fig 2, B shows the resulting curve indicating a linear relationship between UTE signal and proton density (R² = 0,987).
Gamma pass rate computations at 3%/3mm provide a mean value of 99.4±0,5% and 99.1±0,5% for the doses computed from UTEtissues and UTEwater respectively. At 2%/2mm, mean values are 97.9± 1.5% and 97.1±1,7% with a significant difference shown by a Wilcoxon test (p≤0,05). The dose differences are negative in most OaRs and TVs for the UTEtissues maps while closer to zero in UTEwater maps (fig 4-5). Globally mean dose values ranged between -4% to 3,7% with σ∈[0,7%; 6%]. Gamma pass rate results indicate a good correspondence of the UTE based dosimetry with the CT based one and the tissue correction appears to improve performance in that regard. Globally ED values in both UTE-based maps were higher than expected leading to a reduced dose deposition as the doses were computed without re-optimization of the treatment plan. A larger study will be conducted to improve reliability of the results as well as the accuracy of the tissue correction on the dosimetry. Also, as UTE signal can be affected by T1-weighting, assessment of its impact in the dosimetry and its correction will be necessary. This study suggests that UTE derived dosimetry maps could have the potential to plan the dose in head and neck cancers only using MRI images although optimizations and verifications are still required. An additional chemical shift encoded scheme in the spiral VIBE UTE could allow fat-water decomposition and avoid the need of CT images for tissue categorical mask computation.
Nils TANNEAU (Lyon), Laura SAYAQUE, Benjamin LEPORQ, Charlène BOUYER, Frank PILLEUL, Vincent GREGOIRE, Olivier BEUF
11:16 - 11:18
#47813 - PG123 Double inversion recovery with controlled signal suppression.
PG123 Double inversion recovery with controlled signal suppression.
Double inversion recovery (DIR) is a magnetic resonance imaging (MRI) technique in which signal from two types of tissue are suppressed, usually cerebrospinal fluid and white matter [1]. The suppression is achieved with two inversion pulses, which are almost always adiabatic pulses, such as hyperbolic secants. In brain regions affected by off resonance due to strong inhomogeneity in the static magnetic field, inversion may be inadequate, resulting in bright signal. We present a 3D DIR scan with inversion pulses that are robust against such inhomogeneities.
Using an optimal control framework [2], we designed preparation pulses that fulfill the following criteria: desired target flip angle of 180 degrees, insensitivity to off-resonance (inhomogeneities in the static magnetic field) over a range of ±5. ppm, target flip angle independent of RF amplitude from 80% to 115% of nominal RF amplitude. These criteria were incorporated directly into the cost functional of an optimal control framework for pulse optimization. Maximum available RF amplitude and phase variation were in compliance with the scanner’s RF coil and specific absorption rate (SAR) limitations. Experimental data with the conventional and the new inversion pulses were acquired and compared at 3 T in a phantom designed to exhibit strong inhomogeneities in the static field, and in human participants (3 healthy volunteers, 2 patients with multiple sclerosis, 1 patient with post-concussive symptoms, 2 participants with asymptomatic white matter hyperintensities). The phantom consisted of a cylinder (diameter = 1.5 cm, length = 9.5 cm) filled with a gadolinium solution (concentration = 0.05 mmol/ml) immersed in a cylindrical phantom (diameter = 13 cm, length = 17 cm). The phantom was placed inside the scanner with the Gd-filled cylinder perpendicular to B0, in order for the paramagnetic solution in the cylinder to create field inhomogeneities. For the phantom scan, only a single inversion pulse was tested in a fluid attenuated inversion recovery (FLAIR) scan, with a TR of 1650 ms, matched to the nulling of the phantom’s water signal. In the participants, DIR images were acquired with sagittal readout in three dimensions with 1 cubic mm isotropic resolution, 1st inversion time (TI1) = 2550 ms, 2nd inversion time TI2 = 470 ms, repetition time (TR) = 5500 ms, effective echo time (TE) = 293 ms. Images were assessed by a neuroradiologist with 42 years of MRI experience. Only the two inversion pulses were replaced in the modified DIR and no other changes to the sequence were made. The new inversion pulse resulted in nearly perfect inversion across the entire phantom, whereas with the conventional pulse bright artifactual signal appeared in areas with inhomogeneous field (Figure 1). In human participants (Figure 1), such bright signal appeared near the paranasal sinuses with the conventional pulse but, was absent with the controlled double inversion recovery scan. Image contrast of lesions in multiple sclerosis (Figure 2) and in white matter hyperintensities were identical for DIR with the optimized pulses and DIR with the conventional pulses. The bright hyperintense artifact in DIR mimics MS lesions and may result in misdiagnosis. The artifact may also overshadow actual pathology in that region. The modification of the DIR sequence requires no hardware modifications or changes to data sampling and reconstruction, which aids clinical adoption of the scan. The improved DIR was validated in a phantom and a small number of patients and healthy volunteers and future work in more conditions will further validate the scan. DIR with robust inversion pulses prevents artifacts caused by incomplete inversion, without altering image contrast of MS lesions and white matter hyperintensities.
Alexander JAFFRAY (Vancouver, Canada), Christina GRAF, Armin RUND, Stefan STEINERBERGER, Anthony TRABOULSEE, David LI, Alexander RAUSCHER
11:18 - 11:20
#47822 - PG124 Quantitative gradient recalled echo (qGRE) with navigator based correction: a test-retest pilot study on healthy controls.
PG124 Quantitative gradient recalled echo (qGRE) with navigator based correction: a test-retest pilot study on healthy controls.
Quantitative Gradient Recalled Echo (qGRE) MRI enables the in vivo estimation of tissue-specific relaxation metrics, such as R2t*, which reflects microstructural integrity and neuronal density. R2t* has been proposed as a sensitive imaging marker for early neurodegeneration,with prior studies demonstrating its value in Alzheimer’s disease and multiple sclerosis (Kothapalli et al., 2022; Zhao et al., 2016). However, its sensitivity to motion and physiological fluctuations limits reproducibility in clinical environments. In this pilot study, we evaluated in a test-retest fashion qGRE derived metrics (R2t*) in healthy subjects and how the use of a navigator-based correction (NAV) can improve estimation reliability with a minor time penalty. We implemented qGRE, aiming to validate its robustness for future longitudinal and clinical applications, therefore we investigate other metrics (R2* and QSM) can be estimated from the same sequence and that can benefit the NAV correction.
Thirty healthy participants (mean age 39.6 ± 6.4 years) underwent two identical qGRE acquisitions on the same day, with repositioning between sessions. The qGRE protocol included a 3D multi-echo gradient echo sequence (10 echoes, TR/TE1/ΔTE 50/4/4 ms, voxel size 1x1x2 mm³), with and without a phase-stabilized navigator inserted before the final echo (Fig. 1). A 3D T1-weighted sequence was acquired for each session of the test-retest (TR/TE: 8.4/3.7 ms, voxel size: 1x1x1 mm3). This navigator corrects for low-frequency B0 field fluctuations and physiological motion (Wen et al., 2015). R2t* maps were calculated using an established qGRE post-processing pipeline, including navigator-based phase correction, multi-echo fitting, and separation of tissue-specific and BOLD-related contributions (Ulrich & Yablonskiy, 2016). R2* maps were obtained using Auto-Regression on Linear Operations (ARLO) (Pei et al. 2015). QSM maps were obtained with Romeo (Dymerska et al. 2021) for phase unwrapping, iLSQR (Li et al. 2015) for background field removal and susceptibility estimation (Fig. 2). Brain parcellation was performed via FastSurfer (Henschel et al., 2020) on the T1 and transferred to the qGRE space by coregistering the first echo to the T1, to obtain ROI-based values. Reproducibility metrics calculated for uncorrected and NAV-corrected metrics included Intraclass Correlation Coefficient (ICC), Bland-Altman (BA) and passing-bablok (PB) analyses. Fig. 2 shows a representative case of the positive impact of NAV, where the analysed metrics (R2* R2t* and QSM) in areas usually subjected to signal loss can be recovered. NAV correction improved the reliability of (R2* R2t* and QSM) measurements across the majority of both cortical and subcortical regions (Fig. 3). BA plots confirmed reduced or comparable bias in each metric and narrower limits of agreement in NAV-corrected data (Fig. 3). PB analysis corroborated the BA by showing an improvement in bias and variability across all the considered metrics in the test-retest when NAV is employed. ICC was highest in supratentorial areas, where navigator correction most effectively mitigated classical susceptibility artifacts that affects tissue interface areas. Cortical surface projections of ICC metric visually demonstrated the improved reproducibility, with NAVcor achieving more consistent estimate of R2t* (78% of ROIs) between the two acquisitions, however it provides a slightly lower improvement of reproducibility in R2* (69%) and QSM (50%) (Fig. 4). This study confirms the test-retest reliability improvement of R2t*, R2* and QSM metrics derived from navigator-corrected qGRE acquisitions in healthy individuals. The navigator implementation successfully compensates for B0 instability and physiological motion, enhancing data consistency even in regions prone to susceptibility effects. Given these improvements, NAV can be considered a crucial component for standardizing qGRE acquisition protocols in clinical and research applications. The different improvement of the ICC metric in R2* and QSM when compared with R2t* might be explained by a higher susceptibility of R2t* to noise, that can be corrected by the navigator. Navigator-based correction substantially enhances the reproducibility of qGRE-derived R2t*,R2* and QSM measures, demonstrating robustness across cortical and subcortical regions in healthy controls. This supports its integration into future clinical and longitudinal imaging protocols focused on microstructural brain assessment.
Marco CASTELLARO (Padova, Italy), Agnese TAMANTI, Giulio FERRAZZI, Teresa MALTEMPO, Roberta MAGLIOZZI, Valentina CAMERA, Alexander SUKSTANSKY, Dmitriy YABLONSKIY, Massimiliano CALABRESE
11:20 - 11:22
#47837 - PG125 About the synergy of servo navigation and PEERS for motion- and frequency-stabilized 3D EPI fMRI.
PG125 About the synergy of servo navigation and PEERS for motion- and frequency-stabilized 3D EPI fMRI.
Three-dimensional echo-planar imaging [1] (3D EPI) offers higher sensitivity and fewer spin history artifacts than 2D EPI [2-4], but makes fMRI time series more prone to dynamic behavior related to motion, physiology and drifts [4-5]. Servo navigation provides a self-calibrating plug-and-play run-time head motion and frequency correction using short (3.2 ms) navigators [6-7]. Recent work showed that frequency precision must be controlled cautiously for effective EPI corrections with high echo times, and phase equalization exploiting repeated shots (PEERS) was proposed, which fine-tunes the frequency estimates leveraging the repetitive EPI data [8]. This abstract explores the mutual benefits of servo navigation and PEERS for effective motion and frequency correction in 3D EPI time series.
Servo navigation requires three basic components for prospective motion correction (PMC) [6]. First, a 3D orbital navigator is inserted into a 3D EPI sequence between slab-selective excitation and EPI readout [8]. Second, a linear model is calibrated on the fly for parameter estimation by a finite differences method [6]. Third, a control mechanism rotates the imaging gradients and shifts the slab for run-time motion updates. The other two navigator shifts, and phase and frequency are corrected in the reconstruction. In this way, shot-wise 3D rigid motion, and global phase and frequency corrections are applied in run-time.
As the frequency precision decreases with shorter navigator duration, EPI frequency corrections become vulnerable to noise propagation, especially for short navigators [8]. PEERS leverages the repetitive structure of fMRI scans to fine-tune the phase and frequency estimates from the EPI data itself as shown in Fig. 1.
Phantom and in-vivo scans with six volunteers have been performed on a 3T Philips Ingenia scanner using a 32-ch head coil. Scans were conducted in accordance to local ethical regulations. Scan parameters were: FOV=220x200x100mm3, 2.5mm isotropic resolution, flip angle=17.2°, TE=30ms, TR=64ms, volume-TR=2s, Rphase=2.2, Rslice=1.8, 300 dynamics. The subjects performed a visuomotor task [8] with and without servo navigation. Two subjects were asked to repeat a motion pattern in separate runs with and without PMC. A phantom [6] was placed on a platform, which was repeatedly moved in and out by 1 mm also with and without PMC. Realignment and coregistration for statistical analysis were done using SPM12. Figure 2 compares the impact of servo navigation (Servo), PEERS and volume realignment in-vivo for one subject without instructed motion. Each method improves temporal SNR (tSNR) individually. Servo and PEERS push performance beyond retrospective realignment and together marginalize the positive impact of realignment. A consistent tSNR improvement over all subjects is confirmed in Ref. [8]. Note that PEERS is a pure reconstruction method, and PEERS off and on are therefore based on the same data, which is not the case for servo navigation.
Figures 3 and 4 compare the tSNR after realignment for phantom and in-vivo scans, respectively, in the presence of motion. In both figures, the tSNR drop from motion (c) is reduced by both Servo and PEERS individually (d-e) and largely mitigated for both methods together (f). Note that the tSNR in (f) always remains below motion-free tSNR with PEERS in (b). In Fig. 3, PEERS remains largely ineffective without motion correction (d) and can only contribute its tSNR impact in combination with servo navigation (f). Servo navigation successfully performs run-time motion correction and maintains the signal correspondences in k-space over time. By this, PEERS is able to compare shots throughout the repetitive scan series and provides beneficial corrections. However, if motion remains uncorrected, PEERS cannot estimate the phase and frequency parameters well as shown in Fig. 3.
In turn, PEERS provides high-precision frequency and phase estimates from the EPI itself, which eases the precision requirements on the navigator. This enables the use of short (3.2 ms) navigators even for high echo times. By matching the shot-wise phases to the reference volume, PEERS harmonizes frequency- and phase-induced fluctuations of the point spread function over time. In conclusion, PEERS and servo navigation synergistically improve upon pure volume realignment by effective inter-shot and intra-shot corrections and, by this, clearly improve tSNR even after volume realignment. Together, the joint motion and frequency corrections stabilize the voxel time series, achieving robust 3D EPI fMRI with short 3.2ms-navigators.
Malte RIEDEL (Schellerten, Germany), Thomas ULRICH, Samuel BIANCHI, Klaas PRUESSMANN
11:22 - 11:24
#48011 - PG126 Open MRI Pipeline for muscle strain calculation.
PG126 Open MRI Pipeline for muscle strain calculation.
In this work, we present a vendor agnostic implementation of a workflow for measuring strain in the forearm muscle induced by NMES. The pipeline encompasses all steps, from data acquisition using an accelerated CINE 4D flow to data reconstruction and analysis. This approach aims to address several challenges in both the quantitative evaluation of neuromuscular diseases and 4D flow data processing. Neuromuscular diseases are classified as rare, and multi-center/multi-scanner studies are necessary to obtain statistically significant sample sizes. Furthermore, although 4D flow has developed towards a quantitative method, the data processing steps are often non-standardized and rarely openly available, hindering reproducibility1.
A 4-point 4D flow sequence2 was implemented in Pypulseq (version 1.4.3) based on a 3D cartesian GRE acquisition, additionally the CINE acquisition was triggered with NMES stimulation of muscles in the forearm of the volunteer, with each contraction cycle lasting 1.5 s.
In order to reduce the total acquisition time the sequence was accelerated with poisson disc undersampling (US factor 9), while keeping a fully sampled center of dimensions 13 by 10 points and elliptical scanning by trimming the edges of kspace (see Figure 1). With the help of Berkley Advanced Reconstruction Toolbox (BART)3, coil sensitivity maps were estimated using the ESPIRiT algorithm (ecalib), and complex data were subsequently reconstructed by incorporating these sensitivity maps to enhance the quality of the images from the raw k-space data.
The dynamic acquisition was tested on n=6 healthy subjects (age 24-31, 5 females, 1 male) on a 3T whole-body scanner (MAGNETOM Prisma, Siemens Healthineers), while contractions were being evoked by NMES in the forearm muscles. A MRI compatible force sensor was used to record the intensity of the force from the evoked contractions during acquisition. A “gradient probing” sequence to map gradient directions to physical coordinates was also implemented to aid in the interpretation of the data from the 4D flow sequence.
The proposed pipeline investigates muscle dynamics by calculating strain tensors from velocity and derived displacement data and it requires a JSON file containing information about the physical direction of the gradients (obtained from the “gradient probing” sequence) and other sequence parameters necessary for the correct calculations of strain values.
The strain eigenvalue calculation involves computing the deformation gradient tensor from 3D displacement fields, then deriving the Eulerian strain tensor and extracting its eigenvalues which represent principal strains in three orthogonal directions (stretching, intermediate, and compression components). Subsequently strain rates are extracted by fitting time-varying strain curves with sigmoid functions to quantify how quickly the tissue deforms during the contractions.
The 4D flow sequence acquisition parameters were as follows: 27 phases, 2 lines per segment, TR=6.7 ms, TE=4.5 ms, FA=10°, resolution=1.5x1.5x1.5 mm³, venc=0.2 s, and acquisition time of 5 minutes.
The sequence, data reconstruction, and analysis pipeline are available at the links below. The undersampling and elliptical scanning (Figure 1) allow the acquisition time to be reduced from 28 minutes (fully sampled) to 5 minutes. The “gradient probing” sequence showed that the X gradient is increasing in the right direction, the Y gradient is anterior-increasing, while the Z gradient is superior-increasing (with respect to HFS patient coordinates).
Figure 2 shows maps of the displacement and first eigenvalue of the strain tensor, where the muscles activated by NMES are clearly distinguishable. The results over the different phases are reported in Figure 3 along with the sigmoid fit used to calculate the buildup rate. The mean build up rate for all the subjects in an roi in the Flexor Digitalis Superficialis was 2.035 s-1.
Finally, Figure 4 shows the combination of the force results obtained with the force sensor during NMES stimulation and displacement along the z direction. It can be observed that the displacement peaks earlier than the force because the mechanical response of the muscle to the contraction precedes the full development of force transmission through the musculoskeletal system, due to slack in the tendon-muscle complex4. Further improvements are needed on the proposed pipeline to improve its robustness and reproducibility. The analysis in this work relied on hand drawn ROIs, while segmentation based on a conventional anatomical acquisition will be implemented for future application, additionally, the use of open and standardised data format such as musclebids is foreseeable. This work introduced a fully open source implementation of a 4Dflow sequence and the accompanying pipeline to analyse the data obtained with the sequence and extract velocity, displacement and strain values from skeletal muscles that have been stimulated with NMES.
Marta Brigid MAGGIONI (Basel, Switzerland), Sabine RÄUBER, Francesco SANTINI
11:24 - 11:26
#47704 - PG127 Reference region B1+ mapping - a convex optimization approach.
PG127 Reference region B1+ mapping - a convex optimization approach.
Variable-flip angle (VFA) T1 mapping is a common approach for rapid T1 measurement [1]. B1+ mapping is essential in order to correct for transmit field inhomogeneities, which confound T1 measurements [2]. Numerous techniques exist for B1+ mapping in vivo [3-5], yet the required additional scanning can sometimes be undesirable (e.g. time constraints or the need for additional breath holds).
Data-driven B1+ correction methods include modeling B1+ as a smooth multiplicative filter on the T1 map [6]. Another method involves using a reference tissue with known T1 to invert the VFA problem and estimate B1+, which has been demonstrated in breast imaging using fat as a reference and linear interpolation to infill water-dominant tissues [7], though it is computationally intensive for large datasets. Here, we instead pose the infilling as a convex optimization problem, leveraging spatial gradient alignment between the corrupted T1 map and the final B1+ map for VFA chemical-shift encoded body MRI.
In a variable flip angle experiment, the measured T1 is roughly quadratic with the ratio of the actual and nominal flip angles. This is shown below in Figure 1.
We can state then that the T1 is proportional to the square of B1+.
T1=a(B1+_actual/B1+_nominal)^2
where a is a proportionality constant with dependence on nominal T1 and other effects. For simplicity, the ratio of actual and nominal B1+ will be referred to as B1+ from hereon.
T1=aB1+^2
(∂T1)/(∂B1+)=aB1+
∂T1=aB1+ * ∂B1+
Since B1+ varies smoothly in space, adjacent points have similar B1+ at sufficiently fine spatial scales. For a single tissue type with uniform nominal T1, local variations in measured T1 are proportional to B1+. This assumes large contiguous regions of uniform T1 and that voxel-to-voxel B1+ variation is small in the acquired T1 map.
∇T1 = a∇B1+
To generalize to any nonlinear variation of B1+ in space, we can assume that the proportionality constant also varies in space.
∇T1=∇a∇B1+
Given known B1+ values using the reference region method previously described, we use the previous gradient relationship to formulate the interpolation step as an optimization problem as shown below in Figure 2.
The problem was then solved as a conic programming optimization problem with CVXPY [8] interfacing the MOSEK commercial solver (mosek.com version 11.0.18).
In vivo validation was performed on a 1.5T scanner (MAGNETOM Sola, Siemens Healthineeers, Forchheim, Germany) in the pelvis of a female volunteer. Reference B1+ maps were acquired with a Turbo-FLASH pulse sequence with a preconditioning pulse [3]. The parameters were as follows: TE = 1.89ms, TR = 12530ms, FA = 8 degrees, receiver bandwidth = 500Hz/pixel, acquired matrix = 192x256x36.
To generate the reference region B1+ estimates, an in-house pulse sequence was used to acquire variable flip angle chemical-shift encoded data [9]. The parameters were as follows: nTE = 6, TE1 = 0.9ms, ΔTE = 1.2ms, TR = 8.73ms, FA = 3 & 14 degrees, receiver bandwidth = 1080Hz/pixel, acquired matrix = 192x256x44, partial Fourier phase encoding = 6/8, partial Fourier slice encoding = 6/8, R = 2x2 CAIPIRINHA.
Reconstruction produced T1 water, T1 fat and proton density fat fraction (PDFF) maps. Reference region B1+ mapping was performed using the T1 fat map with fat dominant tissues assumed to have a nominal T1 of 280ms. The B1+ estimates from this were then used as the known B1+ for the convex optimization problem.
Comparison was made between the method of Sung et al. using linear interpolation with Delaunay triangulation (griddata algorithm in scipy version 1.15.2) and the proposed method by measurement of root mean square error with the reference. Reconstruction times of the B1+ maps for the proposed method and with linear interpolation for 5 slices surrounding isocentre were 26s and 26min 31s respectively. Reconstructions from the proposed method are shown with varying regularisation weights in the grid below in Figure 3.
Increasing λ results in increasing bleed-through of the T1 map into the resultant B1+ map, whilst increasing μ results in tighter agreement with the reference region data.
The combination of μ = 1, λ = 0.005 produced the smallest root mean square error with the ground truth data (RMSE = 0.0486) and was marginally better than the linear interpolation approach (RMSE = 0.0551). Difference images are shown below in Figure 4. We present an optimization approach for interpolating B1+ from reference region data, achieving over 60× speedup compared to a Python-based linear interpolation routine and improved accuracy. Both methods showed large errors near subcutaneous fat boundaries, where fat T1 variability and tissue transitions may affect accuracy. We demonstrate that convex optimization provides a compelling alternative for B1+ map reconstruction from reference region data. Further validation in broader anatomical contexts and patient cohorts is warranted.
Yassine N. AZMA (London, United Kingdom), Pete J. LALLY, David J. COLLINS, Nina TUNARIU, Dow-Mu KOH, Christina MESSIOU, Geoff CHARLES-EDWARDS, Christina TRIANTAFYLLOU, Neal K. BANGERTER, Jessica M. WINFIELD
11:26 - 11:28
#47775 - PG128 KISME: Kernel-based Incoherent Sampling of Multi-Echo data for the mitigation of physiological noise in relaxometry data.
PG128 KISME: Kernel-based Incoherent Sampling of Multi-Echo data for the mitigation of physiological noise in relaxometry data.
In MRI, human physiology (e.g. cardiac and CSF pulsation, breathing and eye movements) induces periodic modulation of the spatially-encoded signal in k-space, producing aliasing artefacts in the resulting images. Reordering the signal encoding (i.e. k-space trajectory) can scramble the fluctuations spatially [1].
Physiological noise also leads to coherent temporal effects in multi-echo gradient echo images [2] where data at each k-space location are conventionally acquired consecutively in time along the echo train. This biases the R2* estimates and reduces their precision [2]. These fluctuations can be made incoherent over echo time at the cost of increased scan time [3].
Here we present a refined method, Kernel-based Incoherent Sampling of Multi-Echo data (KISME) in which incoherent sampling is extended to the phase-encoded k-space plane, across echo times. Local reordering in this 3D space scrambles temporal signal fluctuations over a defined time window without increasing scan time. The k-space step size is minimised to avoid artefacts due to eddy currents. We validate the method qualitatively at 7T and quantitatively at 3T.
KISME divides k-space into a number of kernels with a specific width in each phase-encoded direction (Fig. 1A). Within each kernel, the k-space traversal is varied each TR by varying the starting k-space location, the intra-kernel route (Fig. 1B), and by adding k-space shifts between consecutive readouts (i.e. TEs, Fig. 1C). Each traversal of the kernel samples all k-space locations but at only one TE per k-space location. Traversals therefore need to be repeated to fully sample the 3D (kx,ky,TE) extent of the kernel.
Each kernel is fully sampled before moving to the next. The starting locations within a kernel are pseudo-randomised to scramble signal fluctuations, while also minimising the step size between consecutive kernels to minimise eddy current related artefacts. This pseudo-randomisation leads to variable time to fully acquire data across TEs at a given k-space location (Fig. 2) mitigating the risk of coherently sampling signal fluctuations.
For qualitative evaluation, 3D multi-echo spoiled gradient echo datasets were acquired on a 7T Siemens Terra. 11 echoes were acquired with TE ranging from 2.30 to 26.10 in 2.38ms intervals. Whole brain coverage, 0.6mm isotropic resolution, an acceleration factor of 2 in each phase-encoded direction and a TR of 31ms led to a scan time of 13.5 minutes. Data were acquired with either standard linear or KISME sampling. R2* maps were computed from the multi-echo data using a log-linear fit and evaluated via visual inspection.
For quantitative evaluation, 3T data were acquired on a Siemens Prisma using a 30 channel receive coil with an integrated head immobilisation system, MrMinMo, to minimise overt participant movement. Either standard linear or KISME sampling were repeated 3 times in randomised order (6 acquisitions total). 8 echoes were acquired with TE ranging from 2.2 to 18.3 in 2.3ms intervals. A TR of 22.5ms and a 6 degree flip angle induced proton density weighting. 1 mm isotropic resolution, whole brain coverage and an acceleration factor of 2 in each phase-encoded direction led to an acquisition time of 3.8 minutes. R2* maps were computed using a restricted maximum likelihood fitting routine [4]. The impact of the sampling scheme was assessed via the precision of the estimated R2* maps across the repeated acquisitions. KISME reduced physiological artefacts, particularly in the cerebellum, hippocampus, brain stem nuclei and posterior to the eyes. At 7T, the KISME images, particularly at later echo times, suffered substantially less artefacts and had much greater detail. This led to sharper anatomical delineation in the R2* maps and the removal of low spatial frequency patches of high/low R2* (Fig. 3).
At 3T, KISME increased the R2* precision (Fig. 4). The median variance was reduced by 22% while its inter-quartile range was reduced by 26%. KISME visibly reduced physiological artefacts at both 3T and 7T. The delineation of fine anatomical details was greatly improved (c.f. u-fibres, Fig. 3). At 3T, over three repetitions, KISME increased the precision of the R2* maps despite the comparatively short maximal echo time and use of the MrMinMo device, both of which increased robustness to physiology-induced and involuntary head motion. KISME limits the maximum step in k-space to mitigate any eddy current-induced artefacts. Its pseudo-randomised nature leads to a distribution of times taken to fully sample a given k-space location ensuring robustness to signal fluctuations above a threshold frequency, allowing the acquisition to be tailored to different imaging scenarios. Crucially, this is all achieved without increasing the acquisition time. KISME scrambles physiology-induced signal fluctuations across both the spatial and temporal dimensions of a multi-echo dataset with no time penalty and improves the reproducibility and definition of R2* maps.
Benjamin JAMES (London, United Kingdom), Quentin RAYNAUD, Frederic DICK, Antoine LUTTI, Martina CALLAGHAN
11:28 - 11:30
#45995 - PG129 Towards fast and accurate quantification of T1, magnetization transfer, and susceptibility at high-resolution in the brain at 3T.
PG129 Towards fast and accurate quantification of T1, magnetization transfer, and susceptibility at high-resolution in the brain at 3T.
Unlike abdominal or cardiac imaging, quantitative MRI (qMRI) has not yet reached clinical maturity for neuroimaging [1]. The known limitations are i. the lack of standardization in the mapping technique leading to high variability of the reported values [2], ii. the fact that confounding effects are not always taken into account in the quantification [3], and iii. The poor spatial resolution or the long acquisition time, which are generally not in line with clinical standards.
In a recent study [4], we aimed to meet these challenges by proposing a fast protocol for joint T1 and macromolecular proton fraction (MPF) mapping using advanced quantitative MT modeling [5, 6]. While this former work compared parallel imaging and compressed sensing techniques, we now intend to extend this approach by adding a deep learning-based reconstruction, as well as extending our multi-echo protocol to extract quantitative magnetic susceptibility (QSM). This not only enables shorter scans and an additionnal biomarker, but also substitute conventional weighted sequences with images derived from our quantitative framework.
Two protocols were compared on a single volunteer at 3T (MAGNETOM Vida, Siemens Healthineers, Germany) using a 64ch receive head coil, with a 3D isotropic 1-mm resolution in sagittal orientation, covering brain and spinal cord down to C7.
Reference protocol (35’): Four sequences were acquired, all with CAIPIRINHA 2×2(1) acceleration and external calibration, except MPRAGE and SWI (R=2, integrated ACS)
-joint T1-qMT: A prototype multi-gradient-echo sequence, MT-prepared with a 12 ms and ±4 kHz sine-modulated Hann-shaped saturation pulse applied at B1,RMSsat of 3.73 µT followed by a 10° readout pulse for MT weighted acquisition, repeated with a single-frequency offset of +100 kHz (unsaturated image) and variable flip angles (VFA) of 6°/10°/25° for 3 additional weighted volume acquisition enabling T1 and MPF joint estimation [6].
-QSM: A gradient-echo sequence with 5 echoes spaced according to the consensus paper (TE1=5ms; ES=6ms) [7], a readout FA=15° and a TR = 33 ms enabling QSM quantification using an integrated research processing pipeline with MEDI algorithm [8].
-MPRAGE: A standard T1w anatomical scan following ADNI recommendation [9] (FA=9°, TI=900ms, TR=2300ms) with a modified rectangular field of view to cover both the brain and the cervical spine as in joint T1-qMT.
-Susceptibility Weighted Imaging (SWI): TE=20ms and TR=33ms.
Accelerated protocol (8’30”): A single execution of the MT-prepared multi-gradient-echo sequence described above and three repetitions of its non-saturated version (VFA), extended to include 6 echoes, were played keeping a constant TR = 30 ms (Fig.1). The non-MTw FA=10° volume was also used for QSM estimation. In addition, this protocol was accelerated with CAIPIRINHA 3×3(1) and reconstructed with a research SENSE-based deep-learning (DL) denoising algorithm [10].
For each protocols a transmit field B1+ map was acquired for correction purposes. Image comparison of T1w, T2*w, quantitative T1, MPF, and QSM maps are shown in Fig. 3. T1w images yielded qualitatively comparable results between protocols. The T2*-weighted image is more contrasted with the reference protocol. The former protocol produces more noise in the center of the brain in consistency with parallel imaging CAIPIRINHA reconstruction. The effect of DL denoising is seen in the second protocol without any a priori contrast deterioration. The quantitative analysis for deep grey matter regions and larger white matter (WM) lobes is shown in Fig. 4. T1 and MPF distributions are narrower for the accelerated protocol, likely reflecting weaker dispersion due to noise within ROIs. Overall, the distributions of both protocols are overlapping for T1, MPF and QSM meaning that the average qMRI metrics are of the same order. Only the T1 values in Caudate show a discrepancy between protocols. Both protocols provided metrics in good agreement. The impact of denoising seen in T1 and MPF narrowed distribution is not seen in all QSM regions. The QSM reference protocol appears to be less affected by noise than the qMT protocol and could be evaluated in future work using MP-PCA noise estimation for both protocols. Besides, there were not many differences between protocols for T1 and MPF estimation compared to QSM.
The T2*w image derived from the accelerated protocol would likely benefit from fine-tuning of the echo combination as compared to the original single-TE SWI. The T1w image from the accelerated protocol can be used for both quantification and anatomical purposes, replacing the traditional MPRAGE. We demonstrated that a multiparametric protocol quantifying QSM, joint T1 and MPF in the brain is feasible within standard clinical acquisition time and spatial resolution without requiring any additional conventional weighted sequences. Further work will focus on optimizing reconstruction parameter for better quantitative and qualitative results.
Anita MASLIAH (Marseille), Lucas SOUSTELLE, Hugo DARY, Kamal ACHALHI, Marcel Dominik NICKEL, Josef PFEUFFER, Maxime GUYE, Stanislas RAPACCHI, Olivier GIRARD, Thomas TROALEN
11:30 - 11:32
#47373 - PG130 Phase graph-based MRI simulation including off-resonant pulse response.
PG130 Phase graph-based MRI simulation including off-resonant pulse response.
Extended phase graph (EPG) simulations [1] or phase-distribution graph (PDG) simulations [2] so far only support on-resonant radio frequency (RF) pulses that act instantaneously. Various aspects of MRI, such as the separation of fat and water signals, however, rely on off-resonant RF pulses. We show here an extension of phase graphs to treat off-resonant RF pulses and allow for simulation of off-resonance effects in MRI. While only block pulses are considered and they are still executed as an instantaneous event, several key MRI features can be simulated by this extension.
In this work we specifically extend the PDG simulation [2] of the MR-zero simulation framework [3] which allows image simulation. For the simulation of MR sequences in PDG, magnetization is decomposed into configuration states, which are described in a complex coordinate system. The action of an RF pulse is implemented via a rotation matrix depending on the RF pulse flip angle and phase, that acts on the magnetization vector. Sodickson and Cory [4] suggest an extension of this formalism to account for RF pulses applied at an off-resonance. The RF pulse amplitude is assumed to be constant. The off-resonance is reflected via a rotation by an effective angle around an effective axis, that is tilted with respect to the B0-axis. The tilt of this axis is determined via the frequency of the B1 field. This results in changes to the amplitudes and phases of the magnetization components compared to an on-resonant RF pulse of the same flip angle [4].
The extended simulation framework is validated via (i) selective pulses for fat-saturation [5], (ii) binomial water excitation [6] and (iii) the B0 and B1 mapping method WASABI [7].
(i) Fat saturation is achieved via a single off-resonant RF pulse (flip angle: 90°, frequency offset: -3.5 ppm, duration: 8 ms) and a dephasing gradient before the imaging sequence.
(ii) Water excitation is done via binomial pulses. For the simulation experiments a 1-2-1 binomial pulse configuration is used to replace the 10 deg excitation pulse in a FLASH readout.
(iii) The WASABI method uses a conventional FLASH readout to sample 30 off-resonant preparations (single off-resonant RF pulse with flip angle: 284 deg, duration: 5 ms) distributed equidistantly across a range of frequency offsets of ±2 ppm.
The pulse schemes for fat saturation and water excitation are shown in Fig. 1A. The brain phantom used for the simulation experiments is built from the data provided in the BrainWeb database [6]. Segmented maps for grey and white matter, as well as CSF are filled with values for the physical tissue parameters. Subcutaneous fat was manually added to the parameter maps. The generated phantom was used to simulate. As the base sequence for the simulation experiments a centric reordered FLASH sequence with imaging parameters TE = 2 ms, TR = 4 ms, FOV = 200 mm × 200 mm, matrix = 100 × 100, flip angle = 10 deg and slice thickness = 5 mm is used. The changes (i)-(iii) are implemented to the sequence respectively. The brain is surrounded by subcutaneous fat (Fig. 1B). Under ideal conditions (constant B0 and B1) the fat signal could be suppressed efficiently by an off-resonant fat saturation pulse, leaving little to none residual fat signal in the image (Fig. 1C). As an alternative method of fat and water signal separation, water-excitation via binomial pulses during the imaging sequence, was simulated. Similarly yielding efficient fat suppression under ideal conditions (Fig. 1E). When B0 and B1 inhomogeneities were introduced, both methods maintained effective fat suppression, although with slightly increased visibility of residual subcutaneous fat. The image acquired using water excitation (Fig. 1F) appeared clearer than that obtained with fat saturation (Fig. 1D). The frequency response of these fat-water separation techniques (Fig. 1G) shows the expected behavior.
Off-resonant excitation was further validated by B0 and B1 field mapping via the WASABI method. The resulting simulated B0 and B1 maps closely matched the corresponding ground truth maps from the simulation phantom (Fig. 2). The implementation of the formalism from Sodickson and Cory [3] into the PDG simulation was shown to correctly simulate the effects of off-resonant RF pulses in different applications. The formalism, however is limited to block pulses, as it assumes a constant pulse amplitude. Other pulse profiles (e.g. Sinc or Gaussian) that violate this assumption are currently not reflected. Furthermore, pulses have to be sufficiently short, such that relaxation effects during the pulse can be neglected. We demonstrated and validated the extension of a phase graph-based MRI simulation with off-resonant pulses. This extension enables the simulation of off-resonance effects of MRI and further increases the accuracy of the simulation of MRI sequences. A generalization of this extension to different pulse profiles and arbitrary pulse durations remains future work.
Felix DIETZ (Erlangen, Germany), Simon WEINMÜLLER, Jonathan ENDRES, Moritz ZAISS
11:32 - 11:34
#46516 - PG131 Investigating ihMT T1D-filtering imaging for characterizing demyelination and inflammation processes in MS lesions.
PG131 Investigating ihMT T1D-filtering imaging for characterizing demyelination and inflammation processes in MS lesions.
The pathological processes involved in multiple sclerosis (MS) lesions, which include demyelination, inflammation, clearance of myelin debris, and induction of glial scar, need to be better characterized. Most of these processes imply cells composed of large and motionally restricted macromolecules (e.g., myelin, macrophages, microglia, astrogliosis). In this context, inhomogeneous magnetization transfer (ihMT) MRI [1,2] may present benefits for macromolecule characterization. ihMT is an MRI technique, weighted by T1D, the dipolar order relaxation time, an endogenous source of contrast driven by slow molecular dynamics and tissue microstructure. As demonstrated in a preclinical study [3,4], long T1D values are mostly associated with myelin, and can be isolated by ihMT high-pass T1D filters (HP); whereas short T1D values are thought to be associated with other macromolecules and can be assessed using ihMT bandpass T1D filters (BP). Consequently, ihMT T1D filters may provide semi-quantitative measurements of both the density of myelin and that of other macromolecules. Here, we investigated ihMT HP and BP T1D filter imaging obtained within a single ihMT sequence in humans at 3 T, and assessed its potential as an indicator of myelin and macromolecule density in MS lesions.
Two healthy controls and two relapsing-remitting MS (RR-MS) patients were scanned on a 3T MAGNETOM Vida (Siemens Healthineers, Erlangen, Germany) using a ~1h protocol approved by the local ethics committee. Acquisitions included: 3D T1-weighted MPRAGE, 3D FLAIR, optimized 3D GRE ihMT for HP/BP T1D filtering (Table 1), and multi-echo GRE (MGE) for QSM reconstruction using MEDI [5]. The ihMT GRE sequence was used to acquire images with multiple saturation conditions (MT+, MT−, MT± (FA, frequency-alternated) with τswitch=1.5 ms, and MT± (CM, cosine-modulated) with τswitch=0 ms), along with a reference image M0.
HP and BP filters were computed as (eq. 1):
HP = ihMTR_FA
BP = ihMTR_CM − ihMTR_FA
where ihMTR = (MT+ + MT− − 2×MT_FA or CM) / M0
Using prior fluorescence microscopy comparisons in mice, linear models were established relating HP and BP signals to myelin density [My] and other macromolecule density [Mm] (eq. 2):
HP = α·[My] + b·[Mm]
BP = α′·[My] + b′·[Mm]
These equations were then inverted to estimate apparent [My] and [Mm] maps from experimental ihMT signals. IhMT HP T1D filters show a very strong GM/WM contrast and an average signal of ~10% in WM. As expected, by their sensitivity to short T1Ds, ihMT BP T1D filters show a weak contrast with a signal of ~3% (Fig. 1). IhMT HP and BP T1D filters demonstrated sensitivity to MS. Used together, they allowed semi-quantitative estimation of the density of myelin and that of other macromolecules in lesions (Figs. 2-3). Their signal profiles across the investigated lesions revealed various levels of demyelination accompanied by an increased density of other macromolecules. Of interest, the macromolecules signal did not always match the QSM signal, indicating a sensitivity of ihMT T1D filters to different types of macromolecules. The semi-quantitative indices [My]* and [Mm]* can be derived from a single MR sequence. The nature of the macromolecules revealed by the [Mm]* signal is not addressed here, but its mismatch with the QSM signal in some lesions may suggest that [Mm]* is sensitive to iron-positive myelin-negative pro-inflammatory macrophages [6-8], and to iron-negative myelin-laden macrophages present in the center of demyelinated lesions [6]. Hence, combining ihMT T1D filtering and QSM may allow for more accurate characterization of the inflammation processes in MS lesions. However, several improvements are essential to make this technique more accurate, more robust and clinically viable:
- reducing sensitivity to motion and transmit-field (B1+) by decreasing the acquisition time, and use of B1-correction strategies, respectively
- increasing the ihMT BP T1D filter signal intensity by optimizing the τswitch value
- improving the accuracy of α, α', β, β' constants (eqs. 1-2), which are currently estimated on the basis of preclinical experiments with non-identical MR sequence variables. This study demonstrates the promising potential of the ihMT T1D filtering technique to refine the characterization of both demyelination and other processes in which different macromolecules are involved (e.g. inflammation) in MS lesions.
Andreea HERTANU (Marseille), Timothy ANDERSON, Lucas SOUSTELLE, Ludovic DE ROCHEFORT, Lauriane PINI, Thomas TROALEN, Jean PELLETIER, Olivier M. GIRARD, Guillaume DUHAMEL
11:34 - 11:36
#46461 - PG132 Quantitative susceptibility mapping in clinical contexts using spatially non-isotropic multi-echo gradient echo data.
PG132 Quantitative susceptibility mapping in clinical contexts using spatially non-isotropic multi-echo gradient echo data.
Quantitative susceptibility mapping (QSM) based on multi echo gradient echo (mGRE) magnetic resonance imaging (MRI) [1] is promising for quantifying iron deposition in the human brain and other organs [2]. In neurological diseases with known iron deposition like Parkinson’s disease, QSM could serve as a valuable biomarker of iron deposition [3]. However, reconstructing quantitative susceptibility parameter maps from mGRE MRI phase information constitutes an ill-posed inverse problem and results depend on image acquisition and postprocessing [4]. Therefore, the Electro-Magnetic Tissue Properties Study Group of the International Society of Magnetic Resonance in Medicine (ISMRM) recently published recommendations for implementing QSM for clinical brain research to promote standardized data acquisition and analysis [5]. In particular, an acquired isotropic spatial resolution of 1 mm is recommended. However, for achieving maximum volume coverage in minimal scan time, clinical QSM acquisition protocols frequently use elongated voxels with higher in plane (typically < 1mm) and lower through-plane resolution (slice thickness > 1 mm) as well as higher acceleration factors and bandwidths (BW).
In this study, we aimed to assess the reliability of already acquired clinical mGRE data with more elongated voxels for QSM. Therefore, we acquired mGRE in ten healthy volunteers with a QSM consensus protocol and a commonly employed optimized clinical protocol, calculated QSM maps using three different evaluation pipelines and compared the resulting parameter maps with respect to artefacts and quantitative susceptibility values in deep gray matter (GM).
Ten healthy subjects (30.9±9.5 y, 4m/6f) were scanned on a 3T Philips Ingenia Elition X scanner (Philips Healthcare, Best, NL) equipped with a 32-ch head coil. Two mGRE protocols were acquired in strictly axial orientation: 1) the spatially isotropic QSM Consensus Protocol; 2) an anisotropic more accelerated protocol with elongated voxels in slice-encoding direction (see Fig.1 for acquisition details). Each 3D mGRE data set was processed using three different QSM reconstruction pipelines: 1) MEDI [6] and 2) STI-iLSQR [7] were applied with default parameters as described before [4]; 3) a prototype implementation of the multi echo complex total-field inversion (mTFI) algorithm [8, 9] was employed as provided by Philips. A systematic assessment of reconstruction quality was performed by visual rating using a 4-point scale (0 = no artifact, …, 3 = worst artifact) according to recommended criteria [10]. Quantitative susceptibility values were extracted from deep GM nuclei affected by Parkinson’s disease, i.e., Substantia Nigra (SN) and Red Nucleus (RN), by transforming the multi-contrast PD25 atlas [11] to each individual subject’s QSM data. For statistical analysis, paired two tailed t-tests were applied to test for statistically significant differences between susceptibility values obtained with different imaging protocols and QSM reconstruction pipelines. Fig.2 shows QSM reconstructions from one subject. While the susceptibility maps obtained from the QSM consensus protocol (top) look similar for mTFI, STI-iLSQR, and MEDI quite some variability can be observed for the QSM reconstructions from the anisotropic more accelerated protocol. Systematic assessment of reconstruction quality revealed stronger artifacts in all categories for the anisotropic clinical compared to the isotropic QSM consensus protocol (see Fig.3). In particular, QSM reconstruction by MEDI failed in two subjects that were excluded from VOI evaluations. Comparison of group average susceptibility values (Fig.4) demonstrated that mTFI, MEDI, and STI-iLSQR yield comparable susceptibility values for the QSM consensus protocol, while there is quite some variability for values obtained from the anisotropic clinical protocol. Notably, the mTFI reconstruction generates high-quality susceptibility maps (Fig.2) with overall minimal artifacts (Fig.3) and stable susceptibility values with both MRI protocols (Fig.4). Quantitative susceptibility maps obtained from the highly accelerated, anisotropic clinical mGRE data seem to be less reliable when reconstructed by MEDI [6] or STI-iLSQR [7] compared to mTFI [8, 9]. This fits with recent work, which recommended spatially isotropic MRI acquisition protocols [5] and demonstrated detrimental effects of low spatial resolution [12] as well as a strong dependence of QSM results on the reconstruction pipeline [4]. As mTFI provided highly comparable results for both investigated MRI protocols, it seems to be a reliable tool for calculating QSM maps from clinically acquired, spatially anisotropic and highly accelerated MRI data. The mTFI method appears to be a promising tool allowing reliable evaluation of highly accelerated mGRE data with non-isotropic spatial resolution that are commonly acquired in clinical contexts.
Dorna HEIDARY (Munich, Germany), Elisa SAKS, Kilian WEISS, Ronja BERG, Benedikt WIESTLER, Jan KIRSCHKE, Dimitrios KARAMPINOS, Jakob MEINEKE, Christine PREIBISCH
|
Salle Major |
|
"Friday 10 October"
Added to your list of favorites
Deleted from your list of favorites
D22
11:00 - 12:30
FT2 LT - Translational MRI beyond anatomy
Functional and physiological imaging
FT2: Cycle of Translation
11:00 - 11:02
#45952 - PG133 Plug’n Play 3D MRSI at 7T with full scanner integration.
PG133 Plug’n Play 3D MRSI at 7T with full scanner integration.
1H-MRSI offers unparalleled insight into brain metabolism without the need to take biological samples, and consequently has application potential in several diseases [1-3]. However, several reasons hold it back from being used in clinical routine. Along with its low sensitivity, the need for expert knowledge in the acquisition, reconstruction, interpretation and evaluation of the results is one of the main obstacles for MRSI for routine clinical application. This expert knowledge includes: Placement of field-of-view (FOV), volume of interest (VOI), B0-Shimming, water-suppression optimization; Image-reconstruction, pre-processing steps, spectral fitting, and absolute concentration calculations; interpretation of metabolic maps, ratio maps, and uncertainty maps. With the recent advances in artificial intelligence, some of these steps may become obsolete or remove the necessity of involving an MRSI expert. Thus, the aim of this study was to implement and briefly compare based on initial insights:
(A) an automated acquisition and scanner-integrated processing pipeline that requires no more than a localizer image and a few mouse clicks to position the FOV/VOI, perform 3D and interactive shimming and start the sequence. The sequence provides both metabolic maps along with corresponding uncertainty (i.e., CRLB) maps within minutes after the acquisition.
(B) our established offline reconstruction, pre-processing, and quantification pipeline running on a dedicated multi-core CPU-server requiring knowledge/experience in programming languages.
We implemented our reconstruction into the ICE framework of our Siemens 7 T scanner. The reconstruction steps included (see Figure 1): Real-time frequency drift correction (via an unlocalized scan, acquired and processed but not used here for this data), coil-compression (from 32 to 10 virtual receive channels to reduce raw data size), averaging of data, channel noise-decorrelation, in-plane field-of-view shift, correcting for chemical shift phase accrual along the ring trajectories, gradient delay correction, k-space density correction to a Hamming filter, in-plane discrete Fourier transform, fast and discrete Fourier transforms. The final data were branched out from ICE to the Siemens framework for image reconstruction environments (FIRE) to perform brain and lipid masking (using iMUSICAL data), L2 lipid regularization (regularization parameter 20E-17), coil combination using iMUSICAL data [4] and voxel-wise spectral fitting. The fitting was implemented as a physics-informed deep auto-encoder which uses a model-based decoder that is very similar to LCModel [5]. After fitting, metabolic, metabolic ratio, and metabolic uncertainty maps are created in ICE. We measured four subjects on our Siemens Magnetom DotPlus 7 T scanner with a 32-channel receive/1-channel transmit coil. The protocol included a 3D concentric ring trajectory (CRT) MRSI sequences [6] with a resolution of 3.4×3.4×3.4 mm³ (matrix 64x64x35, 300ms readout, TR 420 to 450, scan time <10 minutes) and spectral bandwidth of 2778 Hz, an MP2RAGE sequence (for offline masking), and a localizer for placing the MRSI sequence. Raw data is processed at our server with 40 physical CPU cores (Intel(R) Xeon(R) Platinum 8280, 2.70GHz), 504 GiB RAM with Matlab and LCModel. While offline processing would take 3 hours (no L2) to 5 hours (and sometimes even twice as much if servers are busy), the online approach after finishing the CRT-MRSI scan takes additionally 4-5 minutes (no L2), 14-18 minutes (with L2) and about 20 minutes if retro-reconstructed (with L2). A feature is that upon demand the reconstruction provides spectra and maps with and without L2 simultaneously. Shown are the metabolites: total choline (tCho), total creatine (tCr), glutamine (Gln), glutamate (Glu), Glu+Gln (Glx), glutathione (GSH), myo-inositol (mIns), total NAA (tNAA=NAA+NAAG) and taurine (Tau). Figure 2 displays various maps of Volunteer 4, Figure 3 shows subsequent brain slices of Volunteers 1 and 2 and Figure 4 (left) demonstrates the effect of L2 versus no L2 regularization of Volunteer 3 as well a comparison of online with offline processing of Volunteer 2 (right). The online-metabolic maps are of high quality and differ from the offline-maps mostly in the cases were L2 was used since BET brain masking is differs due to different inputs (iMUSICAL vs. MP2RAGE). This results ultimately in different lipid masks. We also observe very slight differences in constrast which propably result from the different fitting methods. As an outlook for the next steps we plan to apply the frequency drift correction and improve and speed up the L2 regularization by deeplearning, for example with WALINET [7]. Automating the MRSI acquisition and reconstruction will greatly improve the applicability of MRSI in clinical routine, as no expert knowledge is necessary. This study will help understand if future full automization (i.e. shimming) is feasible with our current methodology.
Lukas HINGERL (Vienna, Austria), Korbinian ECKSTEIN, Bernhard STRASSER, Aaron OSBURG, Stanislav MOTYKA, Amir SHAMAEI, Philipp LAZEN, Alireza VASFI, Anna DUGUID, Wolfgang BOGNER
11:02 - 11:04
#47881 - PG134 Hyperpolarized 129Xe MRI on a clinical scanner using commercially available cryogen free dDNP polarizer.
PG134 Hyperpolarized 129Xe MRI on a clinical scanner using commercially available cryogen free dDNP polarizer.
Hyperpolarized (HP) 129Xe is an excellent NMR probe for porous media, proteins and tissues [1]. Most importantly, the research and technological development in the past two decades have explored HP 129Xe MRI as a diagnostic tool for diseases related to lung ventilation [2][3][4], reaching FDA approval in 2022 [5]. Also, its ability to penetrate the blood brain barrier proved useful for brain perfusion [6][7]. 129Xe is routinely hyperpolarized using spin exchange optical pumping (SEOP), a very efficient technique, but limited to noble gases [8][9]. On the other hand, Dynamic Nuclear Polarization (DNP) is inherently universal for all NMR active nuclei, and its use is growing to a clinical level for metabolic MRI [10].
The feasibility of hyperpolarizing 129Xe with DNP (xenon DNP), has been demonstrated [11][12][13][14]. The complexity of the solid state mixture preparation led to variable and limited polarization [14]. Simplifying the sample preparation process made xenon DNP more user-friendly and achieved solid-state polarization levels comparable to SEOP [15]. Nevertheless, making xenon DNP readily available for preclinical studies, entails developing a robust and reliable protocol for gas sublimation after dissolution. This work focuses on translating proof-of-principle DNP HP xenon gas [14] to a procedure implementable on a commercially available polarizer.
Xenon DNP sample preparation:
In addition to natural abundance xenon (26% 129Xe), the DNP samples consist of trityl radical (Finland acid) and 2-methylpropan-1-ol. All samples had ~30 mL xenon, 30 mM radical concentration and total volumes 300 µL or 500 µL. The gas admixture into the solvent follows methods described in our previous work [8]. DNP happens at 6.7T and 1.2 K on a SpinAligner polarizer (Polarize ApS, Denmark).
Xenon sublimation:
The buffer volume and push time were adjusted to the custom built sublimation equipment (i.e. liquid/gas phase separator plus a set of valves and tubing) and a TEDLAR bag used to collect the gas (0.6L Tedlar(R) PLV Gas Sampling Bag, Sigma-Aldrich) in order to fully dissolve the sample while at same time avoid liquid residuals in the TEDLAR bag (Figure 1 C and D). The xenon DNP was compared to HP xenon gas produced on a clinical SEOP polarizer (POLARIS, University of Sheffield) with isotopically enriched 129Xe gas (86%) (Linde Gas A/S, Denmark). The gas samples were measured with a dedicated transmit/receive 129Xe 8-channel vest coil (JDcoils, Germany), in a 3T MRI scanner (Signa, GE Healthcare). Increasing the solvent volume while keeping the same amount of gas in the sample improved solid state polarization by 2.5 times (Fig 2A). The buildup times for both volumes are in the range of 1100-1300 s (Fig 2A), but without frequency modulation the buildup time increases to 1800 s (Fig 2B). Comparatively, SEOP can repeatedly cycle hyperpolarized doses of HP 129Xe every 18 minutes. Microwave frequency modulation also notably improves the polarization level by a factor 1.5 (Fig 2B).
Magnetization values were measured on TEDLAR bags in the GE scanner with non-localised MRS of 30° pulses 2 s apart. The relaxation times T1 from SEOP and DNP were 17.4 ± 1.2 s and 20.1 ± 3.1 s (Fig 3). Using standard ventilation imaging protocol we qualitatively assess the B1 homogeneity of the coil. The images with two TEDLAR bags were first with HP xenon from the same SEOP batch (Fig 4A), then with one xenon DNP and one SEOP bag concurrently (Fig 4B).
Coil sensitivity affects the signal intensity, as seen on the dual SEOP bags image (Fig 4A): the top bag’s marker has a SNR of 13.4, and the bottom bag has SNR of 9.5. The same marker distance in the DNP/SEOP image (Fig 4B) have SNR 2.8 and 6.8. The T1 values are consistent with our previous measurement of T1=18 s at a benchtop NMR Spectrometer (SpinSolve, Magritek, Germany) [15].
Adjusting the SNR values (Fig 4) for the B1 sensitivity yields DNP=2.8/SEOP=9.7. However, this SNR difference (a factor 3.4) is almost entirely explained by the difference in enrichment (factor 3.3) between the SEOP prepared batch (86% 129Xe) and the DNP prepared batch (26% 129Xe). The bags have different shapes and proximities to the coil in the two experiments, making an exact marker match impossible. However, slight differences in marker position in the high-intensity areas only give minor differences in the SNR comparison. Using enriched 129Xe in the DNP preparation will lead to equivalent polarization and SNR between the two hyperpolarization techniques. The implementation of xenon DNP at a commercial polarizer, including the sublimation equipment, yielded hyperpolarized xenon gas with an SNR in the range of the conventional SEOP hyperpolarization method. This makes DNP a potential alternative to hyperpolarize 129Xe in facilities without access to SEOP.
Emma WISTRÖM (Lausanne, Switzerland), Jean-Noël HYACINTHE, Esben SOVSO SZOCSKA HANSEN, Michael VAEGGEMOSE, Rolf GRUETTER, Christoffer LAUSTSEN, Andrea CAPOZZI
11:04 - 11:06
#47635 - PG135 To Pool or Not to Pool? Comparability of Multi-Protocol Ultra-High-Resolution qMRI in Healthy Brain Aging.
PG135 To Pool or Not to Pool? Comparability of Multi-Protocol Ultra-High-Resolution qMRI in Healthy Brain Aging.
High resolution imaging associated with ultra-high field (UHF) MRI provides promising advances for brain imaging. A particularly powerful UHF tool is quantitative MRI (qMRI) which aims to remove the protocol dependency by measuring parameters related to the biophysical properties of the tissues.
Few studies have employed large cohorts to infer age-related changes of UHF qMRI parameters [1–4]. A limitation of existing studies is that each has been restricted to its own scan protocol. This constrains further expansion of the cohorts that could be done by pooling multiple datasets.
We combine an open UHF qMRI dataset [1] with locally-acquired data and explore how pooling affects the observed age dependencies, and which biases occur between protocols. We focus on small subcortical structures as their delineation is almost exclusive to UHF MRI.
The sample comprised three cohorts of healthy subjects from different sources referred as AHEAD, sTx-MPM and pTx-MPM. The AHEAD data came from an openly available dataset [1] previously acquired on a Philips Achieva 7T. The other two cohorts’ data sources were the studies conducted at local site on a 7T Siemens Terra. The total dataset comprised 223 subjects (128F, 95M, mean 44.9 y).
The AHEAD study (60F, 45M, mean 42.4 y) used a 1-Tx/32-Rx coil and an MP2RAGEME acquisition [5] with two inversion times (TI1/TI2 670.0/3675.4 ms), 4 echoes (TE of 3.0/11.5/19.0/28.5 ms), and SENSE acceleration with R=2 in one direction. No B1 mapping was performed.
The sTx-MPM study (53F, 32M, mean 48.6 y) employed a 1-Tx/32-Rx coil and an MPM protocol [6] with three (T1-, PD-, MT-weighted) whole-brain 3D FLASH multi-echo sequences (TR 19.5 ms, FA PDw/MTw/T1w 5/5/20°, 6 echoes for PDw and T1w, TE 2.3 to 14.2 ms, and same first 4 echoes for MTw, GRAPPA acceleration in two directions with R=2 in each, and a 4 ms, 140° Gaussian MT-pulse, 2 kHz off-resonance. SE-STE-EPI was used for B1 mapping [7].
The pTx-MPM study (15F, 18M, mean 43.4 y) adhered to a similar protocol, but used a 8-Tx/32-Rx coil, kt-points excitation pulses [8], a 4 ms, 130° Gaussian-shaped MT-pulse, 3 kHz off-resonance, and AFI based B1 mapping [9].
The AHEAD pipeline comprised the LCPCA denoising [10], standard MP2RAGE calculation of the T1 [11], single-exponential fitting of the echo decay for the R2*, and TGV-QSM [12]. The output maps (R1, R2*, and QSM) were then used for the Multi-contrast Anatomical Subcortical Structures Parcellation (MASSP) [13] to provide the ROI for statistical analyses.
The data in sTx-MPM and pTx-MPM cohorts were processed through similar pipelines comprising LCPCA denoising, hMRI-toolbox [14] for quantitative parameter calculation [14], and MASSP. Potential errors were assessed via calculating the number of non-unique R2* values and entropy [15] in the ROI.
Linear models were built for each of the median qMRI metrics in each subcortical ROI with age, age2, and sex being the regressors [1–3]. Significance of the observed age dependency, protocol-related bias or age-protocol interaction was determined at a false discovery rate-corrected [16] threshold of 0.05. High resolution (0.6 mm) maps of qMRI parameters (R2*, PD, and R1) were obtained, allowing parcellation and ROI volume calculation (Fig 1). Quality assurance has suggested excluding the ventricles and few subjects per ROI from further analyses (Fig 2).
The GLM analyses showed significant age and age2 dependency in R1, R2* and subcortical ROI volume for most ROIs. Most of the models indicated significance in protocol-related variable and interactions between protocol- and age- related variables (Fig 3). Age-related changes in the pooled dataset exhibit the inverted U-shape dependence in all qMRI metrics, which arises from age-related myelin loss and iron accumulation in the brain tissue [17].
Comparing AHEAD and local data suggests compatibility of age-related profiles for R2* and ROI volume measurements, having the lowest number of significant age-protocol interactions, and less so for the R1 measurements. Generally, the age dependencies (i.e., the GLM slopes) stay consistent, but the absolute values (i.e., the GLM intercepts) vary across protocols.
R1 was strongly (up to 31% difference) affected by the protocol, potentially due to difference in B1 or bias in B1 measurements [14]. The B1 mapping exhibited systematic B1 difference (Fig 4) in average B1 between sTx and pTx protocols, which could be caused by biases in AFI and SE-STE-EPI B1 mapping [18], resulting in systematic R1 difference. The R2* calculation reduced dependency on B1 mapping can explain it exhibiting less biases and interactions in the pooled dataset, making it stable across UHF qMRI protocols. Pooling UHF qMRI datasets reveals inconsistency in absolute values of qMRI metrics across different protocols due to biases in B1 mapping. R2* and ROI volume display more inter-protocol stability, but R1 can also be used for determining age-related dependencies if protocol difference is accounted for.
Mikhail ZUBKOV (Liege, Belgium), Kerrin PINE, Bazin PIERRE-LOUIS, Puneet TALWAR, Nasrin MORTAZAVI, Solène DAUBY, Chloé GERON, Elise BECKERS, Laurent LAMALLE, Christophe PHILLIPS, Fabienne COLLETTE, Pierre MAQUET, Emilie LOMMERS, Anneke ALKEMADE, Nikolaus WEISKOPF, Evgeniya KIRILINA, Gilles VANDEWALLE
11:06 - 11:08
#47662 - PG136 Automatic radiology assessment of lumbar degenerative diseases.
PG136 Automatic radiology assessment of lumbar degenerative diseases.
Low back pain is the leading cause of disability worldwide, affecting over 619 million people according to the World Health Organization [1]. Despite this, current assessments largely rely on qualitative visual inspection of lumbar MRI scans, which is time-consuming and suffers from inter-rater variability [2]. Classifying the severity (normal/mild, moderate, and severe) of common lumbar degenerative diseases including left/right neural foraminal stenosis (NFN), spinal canal stenosis (SCS), and left/right subarticular stenosis (SAS) remains difficult. While prior work has proposed solutions to these tasks [3–5], these approaches typically require training a dedicated segmentation model for each pathology to extract regions of interest (ROIs), limiting their use. We propose three severity classification models for these diseases as part of the RSNA 2024 Lumbar Spine Degenerative Classification Kaggle Challenge [6]. Our approach is based on a novel preprocessing pipeline that eliminates the need to train specialized segmentation models for ROI extraction, resulting in a more efficient and generalizable solution.
We developed two deep learning based Multiple Instance Learning (MIL) [7] models (described in figure 1) and one ResNet50 model [8] to predict disease severity for the five lumbar intervertebral discs, resulting in 25 predictions per patient (5 vertebral levels per disease: NFN_R, NFN_L, SAS_R, SAS_L, SCS). These models were trained on the RSNA challenge dataset, which includes nearly 2,000 patients with three MRI sequences—sagittal T1w, sagittal T2w, and axial T2w—collected from eight sites across five continents. The dataset was annotated by ASNR radiologists.
The analysis pipeline is summarized in figure 2. Training the models required robust preprocessing to extract meaningful ROIs. We used the TotalSpineSeg [9] model to automatically segment intervertebral discs on sagittal scans. These segmentations were then aligned with axial acquisitions using the Spinal Cord Toolbox [10]. This process enabled accurate extraction of 3D ROIs centered on the discs, leveraging the volumetric nature of MRI data and ensuring spatial consistency across sequences.
Each of the three architectures was trained to predict a specific condition. The models operate at the disc level, processing a single disc ROI at a time. For prediction, the SCS and SAS models utilized the MIL architecture and the axial T2w sequences of each patient, while the NFN model utilized the ResNet50 architecture and sagittal T1w sequences. All models were optimized using the challenge-provided weighted cross-entropy loss function. The outputs from each model are then aggregated to produce severity scores for each pathology across each lumbar level.
Two features of the analysis pipeline were evaluated: (i) The accuracy of ROI selection based on the automatic spine segmentation and (ii) the performance of the model’s predictions. For ROI validation, we used radiologist-provided annotation points to verify whether they were included within the extracted ROIs. This evaluation was only conducted for NFN, as the annotations for SAS were provided on sagittal T2 images, whereas our pipeline used axial T2 scans. As well, the SCS annotations were on axial DICOM images, but the NIFTI conversion process changed the structure by separating the multiple acquisition, preventing their use in validation.
The quality of the model predictions was evaluated using the official Kaggle challenge metric. Our complete pipeline was executed on Kaggle’s servers against a hidden test set, and performance was quantified based on the resulting cross-entropy loss. Our pipeline got an accuracy of 0.97 for NFN ROI extraction.
Our models achieved a score of 0.509 on kaggle, placing us in the top 100 out of 1,875 teams. For reference, the top-performing team achieved a score of 0.389. Our 3D ROI extraction pipeline based on TotalSpineSeg proved to be robust and highly effective. This approach generalizes well and could be applied to other use cases without additional annotations or training, making it a robust and reusable tool in medical imaging workflows.
Our model’s prediction score reached 100 out of 1,875 participants, without requiring post-hoc adjustments on the hidden test set (e.g., manually scaling class probabilities to improve leaderboard scores, something commonly done by top teams on Kaggle challenges). Future work will involve moving beyond challenge-specific metrics to evaluate our approach more comprehensively, using ROC curves and other clinically relevant performance indicators. Our approach demonstrates that accurate and generalizable 3D ROI extraction is feasible without model training or manual annotations. By leveraging anatomical structure and spatial priors, we enable more robust medical image analysis. Future evaluations using ROC curves will offer a more transparent and clinically relevant evaluation of our pipeline's strengths.
Thomas DAGONNEAU (Montréal, Canada), Abel SALMONA, Nathan MOLINIER, Julien COHEN-ADAD
11:08 - 11:10
#46148 - PG137 Comparison of Capabilities for Regional Perfusion and Functional Loss Evaluations among ECG- and PPG-Gated PREFUL MRIs and Dynamic CE-Perfusion MRI.
PG137 Comparison of Capabilities for Regional Perfusion and Functional Loss Evaluations among ECG- and PPG-Gated PREFUL MRIs and Dynamic CE-Perfusion MRI.
Pulmonary functional MRI has been developed and tested by dynamic contras-enhanced (CE-) first-pass perfusion MRI, non-CE-perfusion MRI, hyperpolarized noble gas MRI and oxygen-enhanced MRI in patients with different pulmonary diseases in the last few decades1-3. Since 2009, Fourier-decomposition MRI or phase-resolved functional lung (PREFUL) MRI with electrocardiogram (ECG) was started to test and tried to clinically set as new pulmonary functional MRI as having the potential to assess regional ventilation and perfusion changes in different lung diseases at same time, although this technique is available in several institutions4-6. Recently, we developed PREFUL MRI by ECG and photoplethysmography (PPG) with Canon Medical Systems Corporation and started to test in routine clinical practice. However, no one has directly compared and quantitatively assessed perfusion-weighted PREFUL MRIs (PW-MRIs) with ECG and PPG with quantitatively assessed dynamic CE-perfusion MRI in thoracic oncologic patients with and without emphysema or pulmonary fibrosis. We hypothesized that PW-MRIs with ECG and PPG as well as dynamic CE-perfusion MRI could evaluate quantitatively regional perfusion and pulmonary functional loss in thoracic oncologic patients underlying different lung conditions. The purpose of this study was to directly compare the potential for regional perfusion and pulmonary functional loss assessments among quantitatively assessed PW-MRIs with ECG- and PPG and dynamic CE-perfusion MRI in thoracic oncology patients with and without emphysema or pulmonary fibrosis.
25 surgical treatment candidates due to pathologically diagnosed lung cancers or mediastinal tumors were prospectively examined thin-section CT, ECG- and PPG-gated PW-MRIs and dynamic CE-perfusion MRI at a 1.5T MR system (Vantage Orian, Canon Medical Systems Corporation, Otawara, Tochigi, Japan) and pulmonary function test including %VC, FEV1% and %DLCO/VA. Then, quantitatively assessed regional perfusion maps were generated by pixel-by-pixel analyses from each MR data by means of our proprietary software provided by Canon Medical Systems. For quantitative evaluation of dynamic CE-perfusion MRI evaluation, dual-input maximum slope model was used in this study. To determine regional perfusion, same region of interests (ROIs) were placed and copied over peripheral and central lung zones in each lung field within both lungs on each quantitative perfusion map at the same proprietary software. Finally, overall perfusion was determined as the averaged value from all ROI measurements in each patient. To determine the relationship of regional perfusion between each method, Pearson’s correlation was performed. To compare mean differences of regional perfusion between each two methods, Student’s t-test were performed. Then, the Bland-Altman analyses were performed to determine the limits of agreements between each two methods. To compare perfusion difference among underlying lung conditions, regional perfusions from each method were compared among normal lung, emphysema and fibrosis by Student’s t-test. To assess pulmonary functional loss evaluation capability on each perfusion method, overall perfusion was correlated with %VC, FEV1% and %DLCO/VA by Pearson’s correlation. Representative cases are shown in Figure 1. There were significant correlations between each method (PW-MRI with ECG vs. PW-MRI with PPG: r=0.74, p<0.0001; PW-MRI with ECG vs. dynamic CE-perfusion MRI: r=0.33, p<0.0001; PW-MRI with PPG vs. dynamic CE-perfusion MRI: r=0.24, p<0.0001) (Figure 2). Mean differences between each PW-MRI and dynamic CE-perfusion MRI were significantly larger than that between both PW-MRIs (p<0.05). The limits of agreements between both PW-MRIs were markedly smaller than those between each PW-MRI and dynamic CE-perfusion MRI (Figure 3). There were significant differences of regional perfusion between normal lung and emphysema or fibrosis on each PW-MRI (p<0.0001), although dynamic CE-perfusion MRI showed significant differences of regional perfusion between emphysema and normal lung or fibrosis (p<0.0001) (Figure 4). When correlated between each overall perfusion and pulmonary functional test results, there were significant positive correlations between pulmonary function test results and overall perfusion determined from PW-MRIs with ECG (%VC: r=0.55, p=0.004; FEV1%: r=0.53, p=0.007) and PPG (%VC: r=0.66, p=0.0003; FEV1%: r=0.54, p=0.005; %DLCO/VA: r=0.4, p=0.04). PW-MRI with ECG and PPG had equal to or better potential than that with dynamic CE-perfusion MRI for regional perfusion and pulmonary functional loss assessments in thoracic oncology patients with different underlying lung conditions.
Yoshiharu OHNO (Toyoake, Japan), Alicia PALOMAR-GARCÍA, Ozaki MASANORI, Bruno TRIAIRE, Kaori YAMAMOTO, Natsuka YAZAWA, Yuichiro SANO, Maiko SHINOHARA, Masato IKEDO, Masao YUI, Takahiro UEDA, Masahiko NOMURA, Takeshi YOSHIKAWA, Daisuke TAKENAKA, Masahiro ENDO, Yoshiyuki OZAWA
11:10 - 11:12
#46372 - PG138 On the effect of anesthesia-driven blood velocity and T₂ changes on labeling efficiency in pCASL measurements in mice.
PG138 On the effect of anesthesia-driven blood velocity and T₂ changes on labeling efficiency in pCASL measurements in mice.
Among arterial spin labeling (ASL) perfusion MRI techniques, pseudo-continuous ASL (pCASL) has emerged as a reliable and non-invasive method for quantifying cerebral blood flow (CBF) [1]. However, accurate CBF quantification depends critically on several physiological and technical factors, including labeling efficiency (IE), which can introduce substantial bias if not taken into account [2].
Optimizing pCASL for preclinical functional imaging presents unique challenges, particularly due to the variety of anesthesia protocols and associated effect on CBF [3]. While it is generally assumed that phase optimization, which is B0 and B1-dependant, can be performed at the beginning and remains stable for the duration of the exam [4], IE may vary as a result of changes in blood velocity and T₂ relaxation time [5]. Notably, increased arterial flow velocity in humans has been associated with reduced IE, thereby affecting sensitivity and complicating quantification efforts [6].
In this study, we examine the impact of different anesthesia and air/O₂ mixtures on IE and CBF in mice, and disentangle the respective contributions of blood velocity and T₂-related changes to IE degradation.
Wild-Type male mice (7-9 months) were scanned in an 11.7T Bruker MRI with a 72-mm quadrature volume coil and a 10-mm single-loop surface coil.
Exp.1 (n=2): To assess anesthesia-induced changes in IE and CBF, anesthesia was initially set to 1.5% isoflurane (iso) in a 1:1 air/O₂ mix, then switched to subcutaneous infusion of medetomidine (med- 0.2 mg/kg/h) with 0.8% iso (transition within 10 min). IE was continuously measured in the carotid arteries (3 mm from the labeling plane, figure 1a-b) before, during, and after the transition (figure 1c), using a ASL-encoded flow-compensated FLASH sequence [7]. pCASL parameters were: B1avg=3.5 µT, Gmax/Gave=90/10 mT/m, labeling duration τ=300 ms, no post-labeling delay (PLD).
Exp.2 (n=3): To investigate the effect of blood T₂ changes on IE, gas mixture was alternated between 1:1 air/O₂ mix and 100% O₂ (figure 2a).
Exp.3 (n=2): To determine whether IE changes were velocity-driven, blood velocity in the carotids was measured at the same location (figure 2b-c), using a phase-contrast MRI sequence (FlowMap, TR/TE=60/3 ms).
Exp.4 (n=4): To examine the impact of anesthesia and gas mix on CBF, pCASL scans were performed (figure 3) using a pCASL-RARE sequence (30 repetitions, TE/TR=22/5000 ms). pCASL parameters matched IE parameters with τ=3000 ms and PLD=300 ms. The imaging plane was set 10mm downstream from the labeling plane. All CBF were computed with a Buxton model [8] and using subject-specific T₁ maps and IE values.
Blood magnetization profiles along the flow direction were simulated [5] with blood T₁=2800 ms [7] and pCASL parameters described below, for a range of blood T₂ values and flow velocities, and corresponding IE were extracted (figure 4). With air/O₂, IE was 0.82±0.03 under iso, and decreased to 0.57±0.05 after switching to med-low iso (figure 1c). Lowering Gmax/Gave to 45/5 mT/m worsened the effect, while increasing to 135/15 mT/m offered no improvement (data not shown). Simulations showed that IE increases with blood T₂ and velocity, but show less dependency to blood velocity within the range of values in this study (figure 4).
Regardless of the gas mixture, transition from iso to med resulted in a decrease in carotid blood velocity from 147±6 mm/s to 91±7 mm/s (mean ± SEM, figure 2b).
With O₂ only, IE did not decrease after med infusion. Switching to air/O₂ caused a drop in IE from 0.82±0.02 to 0.55±0.07, which was reversed upon reintroduction of O₂ only (figure 2a). A ~40% decrease in CBF was observed under the med+air/O₂ condition. However, with 100% O₂, no significant change in CBF was observed between anesthesia conditions (figure 3). The observed decrease in IE is most likely due to a combination of blood T₂ shortening and, to a lesser extent, to blood velocity decrease, as supported by simulation and experimental data (Exp.2-3). T₂ changes are likely driven by reduced blood oxygenation under the med+air/O₂ condition [9]. However, administration of 100% O₂ appears to restore IE, likely due to an increase in blood oxygenation and, consequently, blood T₂.
Yet, the CBF increase under med+100% O₂ was unexpected as appropriate account of the IE value should prevent any bias. While the lower CBF value observed with med+air/O₂ compared to iso only aligns with previously reports [3], hyperoxia was reported to induce no change in global CBF [10]. We cannot exclude that other factors may affect quantification in these conditions. These results emphasize that changes in blood velocity and oxygenation due to anesthesia and carrier gas can strongly affect IE, and thus ASL signal to noise and absolute CBF quantification. Although less critical for comparative studies employing consistent anesthetic protocols, awareness of these effects remains essential to minimize bias and ensure robust interpretation.
Sophie MALAQUIN (Paris), Lydiane HIRSCHLER, Celine BALIGAND
11:12 - 11:14
#47398 - PG139 RARE-Readout pCASL for Quantitative Functional CBF Imaging in Rats at 11.7T: Overcoming EPI Artifacts in Post-Surgical Somatosensory Cortex.
PG139 RARE-Readout pCASL for Quantitative Functional CBF Imaging in Rats at 11.7T: Overcoming EPI Artifacts in Post-Surgical Somatosensory Cortex.
Somatosensory-evoked fMRI studies are traditionally conducted using an EPI readout for fast acquisition and high T2* contrast. In rodent models of brain pathology, stereotaxic surgery is often required for the delivery of viral vectors, pharmacological agents, or the implantation of devices, which can make echo-planar imaging (EPI) particularly challenging. Indeed, EPI images are strongly affected by the large magnetic susceptibility differences surrounding scare tissue, or implants. In these conditions, and in particular at higher magnetic field, strong artifacts appear that may affect the evaluation of the local functional response [1-3]. Moreover, while classic BOLD fMRI is very sensitive to vascular reactivity, it is not quantitative. Its analysis relies on statistical parametric mapping, thresholding and report the “activated” pixels count [4]. On the other hand, arterial spin labeling (ASL) MRI is a quantitative approach providing a measure of cerebral blood flow (CBF). Only a few studies have applied functional ASL MRI in rodent [5,6], and an EPI readout was used. In this work, we combined a pCASL module [7] with fast spin echo readout (RARE). We show that it can be used in animals after they underwent surgery in the somatosensory cortex to study the CBF response to electrical paw stimulation.
Four female Wistar rats received bilateral stereotaxic injections of an AAV expressing mCherry (2/DJ, CBA promoter) into two somatosensory cortical sites (S1HL/S1FL), serving as a control to reproduce typical AAV delivery procedures, without any functional gene modulation. Four weeks post-surgery, rats were scanned on an 11.7 T Bruker system with a 72 mm volume coil and a ¹H surface coil, under a mix of medetomidine (0.3 mg.kg⁻¹.h⁻¹) and 0.5 % isoflurane delivered in 100% O2. The pCASL-RARE sequence was written in Paravision 6.1. After global shimming, a series of pCASL pairs of images were acquired with 25 phase sweep steps (-15 to 360°) to determine interpulse phase correction (TR/TE = 2000/15 ms; 4 mm slice; 250×250 µm²; labeling time τ = 1500 ms; PLD = 200 ms; B1avg = 3.5 µT; Gmax/Gave = 90/10) [7]. Inversion efficiency (IE) was measured 5 mm downstream (TR/TE = 225/3.5 ms; 1 mm slice; τ = 200 ms). Unilateral fore- and hind-limb stimulation was used for all functional scans (1.75 mA, 50 ms pulses, 5 Hz; block paradigm 30 s ON/30 s OFF). Functional pCASL-RARE label and control images were alternately acquired for 2 blocks of 10 minutes, with an IE acquisition in between blocks (TR/TE = 5000/21 ms; τ = 2000 ms; PLD = 300 ms; 2 mm slice; resolution = 250×250 µm²). Inversion-recovery RARE images were used to compute T1 maps. Isoflurane was then switched off. Ten minutes later, BOLD fMRI was acquired first with GE-EPI (TR/TE = 1000/15 ms; 1 shot; 128×64 matrix; 14 slices; thickness 0,75 mm; FA = 60°, 10 min) then with SE-EPI (TR/TE = 1000/18 ms; 2 shots; 10 min). Data were analyzed in Matlab. All CBF were computed with a Buxton model [8] and subject-specific IE values. BOLD fMRI data were processed in SPM12, with motion correction followed by GLM analysis (p < 0,001 for GE-EPI and p < 0,05 for SE-EPI). As expected, GE and SE-EPI BOLD images were strongly impacted by the injection procedure (Fig 1). Two-segment SE-EPI mitigated the geometric distortion and improved images quality, however decreasing sensitivity to BOLD contrast compared to GE-EPI (119 ± 59 activated voxels for GE-EPI vs. 41 ± 32 for SE-EPI). GE-BOLD data showed that the functional vascular response spanned over 3 mm in the slice direction, supporting our choice of a 2 mm slice thickness for pCASL-RARE acquisitions. Surgery-induced artifacts were fully abolished in the RARE images (Fig 1). CBF maps averaged over the “ON” periods showed an increased perfusion localized in the contralateral somatosensory cortex compared to the “OFF” period (133 ± 21 vs. 115 ± 17 mL.100g-1.min-1, mean ± S.E.M, Fig 2), i.e. ∆CBF = 15.4 ± 1.6 %. These preliminary results show the feasibility of functional pCASL-RARE. Despite variability in cortical CBF in the OFF condition in our small animal group, ∆CBF fell within a narrow range (12-20%). This value was consistent with previous reports [5,6], although direct comparison is limited due to differences in anesthesia protocols. It is possible that our current manual ROI selection included voxels outside the “activated area”, thereby underestimating ∆CBF. More work is underway to refine post-processing, confirm the extent of the CBF increase in our conditions, and establish reproducibility in a larger group of animals. pCASL combined with a RARE readout provided distortion-free quantification of the vascular response induced by electrical paw stimulation, even post-surgery in the S1 cortex. This approach could be used in other cortical surgery models or fiber implants for functional studies at high magnetic field and is a quantitative alternative to BOLD contrast to assess the vascular response to neural activation.
Cameron HERY (Paris), Sophie MALAQUIN, Lydiane HIRSCHLER, Celine BALIGAND
11:14 - 11:16
#47478 - PG140 Superselective Arterial Spin Labelling revealed chronic perfusion alterations in patients with internal carotid artery stenosis.
PG140 Superselective Arterial Spin Labelling revealed chronic perfusion alterations in patients with internal carotid artery stenosis.
Cerebrovascular diseases (CVD) are a major health issue in developed countries, which are especially associated with increased risks of ischemic stroke [1, 2].
While CVD typically induce narrowing of the brain feeding arteries and hypoperfusion in the dependent vascular territories, there are protective pathways such as collateral blood flow over the circle of Willis [3, 4]. Therefore, treatment decisions require a high degree of individualized diagnosis to assess the perfusion status correctly. Currently, vessel-selective imaging is clinically performed using catheter-based digital subtraction angiography, which, however, comes with intervention risks and the need for hospitalization [5].
A non-invasive alternative for individual mapping of vascular perfusion territories is superselective pseudocontinuous Arterial Spin Labelling (ss-pCASL), which allows selective labelling of specific brain feeding arteries [6, 7].
While previous studies focused on qualitative description of perfusion alterations [7-9] or reported inconclusive findings regarding the shift of territories [10], the purpose of this work was to implement a quantitative assessment of perfusion territory shifts based on ss-pCASL. Therefore, we acquired data in two patient groups: patients with atherosclerosis-induced internal carotid artery stenosis and younger patients with moyamoya disease. We hypothesized that asymptomatic a-ICAS may induce perfusion territory shifts, and that a similar method could be applied for moyamoya disease.
We acquired data in 23 subjects on a 3T MRI (Ingenia Elition X, Philips, Netherlands), from which we included 8 patients with atherosclerosis-induced asymptomatic, unilateral and high-grade ICAS (a-ICAS, 69.8±6.2y, 5f), 3 moyamoya patients (31.7±3.7y, 3f), and age-matched healthy controls (HC, n=20, 69.2±5.8y, 12f); 3 subjects didn’t meet the inclusion criteria. The multi-parametric MRI protocol (Fig.1) included ss-pCASL of the left and right internal carotid artery, time of flight angiography and structural T1w-MRI.
Image processing was based on SPM12 [11] and MATLAB (v2021b, The MathWorks Inc., USA). Based on ss-pCASL perfusion maps, we semi-automatically segmented vascular perfusion territories using Vinci (v5.06, MPI, Germany). From those territory masks, we derived three quantitative parameters:
1. fractional volume (in comparison to whole brain volume)
2. territorial shift (volume fraction comprised in the opposite hemisphere)
3. overlap with an atlas of vascular territories [12] (DICE coefficient, 0 Example data (Fig.2) of a right-sided a-ICAS patient (patient 1, left) show a marked shift in perfusion, where hypoperfused anterior regions of the ipsilateral hemisphere (A) are supplied from the contralateral side (D). Perfusion territory segmentations are shown (B, E), with the shifted region indicated in red (E). Contralateral hemispheres show a larger overlap with the atlas (F vs C). Similarly, results can be seen in data of a moyamoya patient (patient 2, right), where hypoperfused regions of the affected side (a) are perfused from the contralateral hemisphere (b).
Statistical evaluations (Fig.3, Tab1.) for a-ICAS show significantly larger volume (A, 31.10±6.20% vs. 13.24±9.00%), shift (B, 5.58±5.55% vs 0.72±1.76%), and overlap (C, DICE,contra=0.67±0.14 vs. DICE,ipsi=0.45±0.25) from contralateral hemispheres, while HCs’ data remain symmetrical. Similar results can be found in moyamoya (Tab.1.). As hypothesized, ss-pCASL-based vascular territory mapping revealed and allowed to quantify shifts of vascular perfusion territories. With respect to a-ICAS this is an interesting finding as the literature reports are mixed: while multiple studies reported shifts in highly stenosed and symptomatic patients [9, 13], other studies found insignificant shifts for asymptomatic patients [10]. Most likely, this was due to the inclusion of lower degrees of stenosis, as higher degrees of stenosis are more likely to be associated with a stronger shift of perfusion territories [13]. This also agrees with findings in a similar cohort, where shifts of border zones of vascular territories were detected based on dynamic susceptibility MRI-based time to peak maps [14, 15].
With respect to moyamoya induced ICAS, our results agree with literature findings from selective MR angiography [16], blood oxygen level dependent (BOLD) MRI and ASL reactivity studies [17, 18], or after revascularization therapy [19]. In conclusion, our results revealed chronic perfusion alterations induced by a-ICAS and moyamoya disease, which manifests as a shift in vascular territories. These shifts can be reliably quantified using ss-pCASL.
Gabriel HOFFMANN (Munich, Germany), Miriam REICHERT, Jens GÖTTLER, Michael HELLE, Lena SCHMITZER, Moritz HERNANDEZ PETZSCHE, Claus ZIMMER, Christine PREIBISCH, Michael KALLMAYER, Kornelia KREISER, Nico SOLLMANN, Hans LIEBL, Stephan KACZMARZ
11:16 - 11:18
#47847 - PG141 Data-driven cerebrovascular reactivity and vascular lag mapping in gliomas with multi-echo BOLD fMRI.
PG141 Data-driven cerebrovascular reactivity and vascular lag mapping in gliomas with multi-echo BOLD fMRI.
Cerebrovascular reactivity (CVR) measures the brain's ability to regulate blood flow in response to variations in arterial CO2 levels [1]. Performing a breath-holding (BH) task while collecting BOLD fMRI data is a simple and non-invasive approach that ideally uses the end-tidal pressure of CO2 (PetCO2) signal to estimate maps of CVR in units of %BOLD/mmHg and vascular delay maps in seconds [2]. These maps can provide clinically relevant in glioma patients to examine neurovascular uncoupling or delineate regions affected by the tumour due to abnormal vasculature [3][4]. However, obtaining reliable PetCO2 recordings is challenging due to task compliance and/or equipment availability[5]. Here, we investigate whether Rapidtide, a data-driven approach for mapping vascular delay using a refined average brain signal, can generate reliable CVR and vascular lag maps when PetCO2 quality is insufficient in glioma patients.
24 glioma patients (28-69 y.o.) with diverse tumour characteristics (Fig. 1) were scanned (3T Siemens PrismaFit, 64-channel head coil) during a BH task, including 8 trials with expirations before and after the apnea [6,7]. MRI data acquisition: ME-fMRI data was acquired with a T2*-weighted gradient-echo multi-echo EPI sequence (TEs=10.6/28.69/46.78/64.87 ms, TR=1.5s, 2.4mm isotropic voxels, SMS=5, GRAPPA=2, PF=6/8, 340 scans). T1-w MPRAGE (pre/post-Gd) and T2-w FLAIR images (voxel size=1mm3) were also acquired. Physiological data acquisition: Exhaled CO2 and O2 levels were recorded via a nasal cannula with an ADInstruments ML206 gas analyzer connected to an MP160 BIOPAC (freq = 40 Hz). PetCO2hrf signal generation: End-tidal CO2 peaks were manually identified using Peakdet [8], linearly interpolated (PetCO2 signal), convolved with the canonical HRF, and downsampled to TR (PetCO2hrf signal). ME-fMRI data preprocessing (AFNI): Volume realignment to the 1st echo single-band reference image was estimated and applied to all echoes. T2*-w echo-combination and ME-ICA with TEDANA [9] with manual evaluation of BOLD-related (accepted) and noise-related (rejected) independent components with RICA [11]. Spatial smoothing (FWHM=2mm) was applied. CVR data analysis: CVR and vascular delay maps were obtained using two methods: Phys2cvr [12] and Rapidtide [14,15]. Phys2CVR: A lagged regression analysis is applied using the PetCO2hrf regressor (61 shifts between -9 to 9 s, i.e. temporal shift=0.3 s), and the realignment parameters and their temporal derivatives, up to 4th-order Legendre polynomials, and the rejected ME-ICA time courses previously orthogonalized to the lagged PetCO2hrf signal and the accepted ME-ICA time courses as nuisance regressors [6]. The bulk shift was estimated via cross-correlation between the PetCO2hrf signal and the average signal of non-tumoral voxels. Rapidtide: Maps were obtained using an equivalent lagged correlation where the regressor of interest is defined as a band-pass filtered (0.009–0.15 Hz) version of the average whole-brain BOLD signal, and “despeckling” using a spatial median filter to correct erroneous delay estimates due to its inherent autocorrelation [5, 14]. Figure 1 provides a qualitative description of the BH task performance (based on CO₂ recordings and respiratory belt), and CVR/delay maps from both methods. Fifteen patients showed good task performance and adequate PetCO₂ signals. Of these, 13 had comparable CVR and delay maps with both methods, showing a decreased CVR and longer delays in tumour regions; two showed decreased CVR with both methods but no longer delays; and the remaining two showed no CVR or delay response with either method. Additionally, five patients exhibited a medium task performance (5/8 valid BH trials), showing decreased CVR in both methods, but only Rapidtide detected longer vascular delays in tumour regions in almost all cases (4 out of 5). Four patients showed poor PetCO₂ recordings despite having good respiratory belt signals (i.e. valid BH performance). In these cases, only Rapidtide captured decreased CVR and lag in tumour regions. Figure 2 presents a representative case with a good PetCO₂ signal, where both methods revealed decreased CVR and prolonged vascular delays in tumour regions. Figures 3 and 4 show cases with poor PetCO₂ signals, where only Rapidtide detected prolonged vascular delays in tumour-affected regions. When PetCO₂ recordings are reliable, Rapidtide produces similar CVR and lag maps to those obtained with phys2cvr, although often yielding smoother and more robust maps. However, when PetCO₂ recordings are missing or of poor quality, Rapidtide becomes a reliable alternative to generate clinically relevant CVR and delay maps without requiring external physiological signals. This study demonstrates the usefulness of Rapidtide, a data-driven lagged correlation method, to yield clinically relevant CVR and vascular lag maps with a feasible BH task in glioma patients, even in the absence or insufficient quality PetCO₂ recordings.
Cristina COMELLA LUENGO (Donostia-San Sebastian, Spain), Lia HOCKE, Stefano MOIA, Santiago GIL ROBLES, Iñigo POMPOSO, Manuel CARREIRAS, Ileana QUIÑONES, Cesar CABALLERO
11:18 - 11:20
#47034 - PG142 Oxygen-glucose index measurements in the rodent brain by 17O-MR at 11.7T.
PG142 Oxygen-glucose index measurements in the rodent brain by 17O-MR at 11.7T.
Understanding imbalances in oxygen and glucose metabolism is essential for exploring brain function and dysfunction. We previously developed a protocol to non-invasively and quantitatively measure the cerebral metabolic rate of oxygen (CMRO₂) by 17O-MRI in mice [1]. A concurrent assessment of glucose consumption is needed to fully capture brain metabolism. The gold standard for measuring cerebral glucose metabolism (CMRglc), 18FDG-PET, suffers from high inter-subject variability in rodents and is incompatible with simultaneous 17O measurements. This study explores an alternative approach based on the detection of 17O-labeled water (H₂¹⁷O) [2] produced during glycolysis (enolase step, Figure 1) following the injection of glucose-6-17O (17O-Glc). We achieved simultaneous monitoring of H₂¹⁷O and 17O-Glc kinetics, and enriched previous quantification model [2] to include glucose transport. In addition, the feasibility of measuring CMRO₂ and CMRglc in a single exam was demonstrated, and the oxygen-glucose index (OGI) was measured.
Experimental protocol: Non-localized 17O MRS was performed on a 11.7T Bruker scanner using a 10 mm-diameter 17O surface coil and a 72mm-diameter 1H-volume coil. Mice were anesthetized with 1.5–1.75% isoflurane and received an intravenous tail vein injection of 17O-Glc dissolved in 0.9% NaCl over 110s (35% enriched, 170μL/min, Nukem). Initial experiments were performed using a 2.5 mg/g dose (n=2), based on a previous report [2]. To mitigate the hyperglycemic conditions, the dose was reduced to 1.25 mg/g (n=3). After global shimming, a series of 17O spectra were acquired with a 6s time resolution (TR= 15ms; 10 μs broad pulse). A subset of 3 mice underwent OGI measurements. After a 5-10 min baseline, mice were transiently delivered 17O2 gas (46% enriched, 50mL/min, Nukem) over 3 min [1], followed by an injection of 17O-Glc 20 min later.
Data processing and modeling: Spectra were processed with Matlab. The injected glucose contributed noticeably to the total MR signal, producing a small peak at 10 ppm from water. To disentangle the contributions of H₂¹⁷O and 17O-Glc, each spectrum was fitted using a Lorentzian model for water and a Gaussian model for glucose. This enabled simultaneous extraction of the time courses of H₂¹⁷O production and 17O-Glc dynamics. The glucose curve was used to estimate the tissue input function (Ctissue) based on an irreversible two-compartment kinetic model (Sokoloff’s). We assumed 5% blood volume and we used the plasma input function (Cplasma) from [2]. Subsequently, a 3-phase model including CMR and flux parameters (KG and KL) [2,3]—was applied to the H₂¹⁷O curve to compute the apparent CMRglc (Figure 2). For OGI data, a 5-phase model was applied (pre-, during and post 17O₂ inhalation, then during and after glucose injection). Normalization to baseline signal assuming [H2O]brain= 16.07 µmol yielded results in µmol/g/min. 17O-Glc signal increased immediately upon injection (Figure 3). Using Sokoloff’s model, Ctissue was successfully recovered, and could be used as an input to fit the 3-phase model to H217O signal (Figure 3B). Despite a reduced glucose signal amplitude, half-dose data were exploitable and yielded CMRglc values consistent with higher dose results. A systematic ~5 min delay in H₂¹⁷O signal increase was observed post glucose injection, similar to that reported by Borowiak et al. [2]. Part of this delay, attributed to tissue uptake, was accounted for by Sokoloff’s model, reducing the delay correction to 2 min. The resulting fit yielded CMRglc = 0.22 ± 0.05 µmol/g/min (n=5). In OGI experiments (Figure 4), the 5-phase model yielded CMRglc = 0.47 ± 0.07 and CMRO2 = 2.26 ± 0.45 µmol/g/min, i.e. OGI = 4.81 ± 0.23 (n=3). Separate processing of CMRO2 and CMRglc data with 3-phase models resulted in lower CMRglc (0.28 ± 0.07 µmol/g/min) but did not affect CMRO2 (2.24 ± 0.45 µmol/g/min). Previous CMRglc reports in rodents, mostly from 18FDG-PET studies [4-6], display variability [0.2 – 0.7 μmol/g/min]. Our results, measured at the enolase step, fall within this range. 18FDG-PET probes hexokinase, therefore providing a theoretical upper limit in the context of our study, due to possible diversion of glucose towards anabolic pathways upstream from enolase (Figure 1). The difference in CMRglc values obtained with the 3-phase and the 5-phase model may result from improved estimation of KL -closely coupled with CMRglc- in the 5-phase model, where KL is jointly constrained by CMRO₂ and CMRglc data. The remaining delay observed after 17O-Glc injection may reflect unmodeled physiological aspects. OGI values were lower than the theoretical value of 6 (complete glucose oxidation), as frequently reported in humans [7]. Here, isoflurane anesthesia, known to enhance brain lactate concentration [8,9], may further increase this decoupling. This study demonstrates the feasibility of extracting and modeling 17O-Glc kinetics, and shows the potential of 17O-MRS for OGI estimation.
Lucie RANNO-CHARRIER, Adélaïde PATOUILLET, Sophie MALAQUIN, Julien VALETTE, Celine BALIGAND (Fontenay-aux-Roses)
11:20 - 11:22
#46639 - PG143 Iron concentration and longitudinal relaxation rate (R1) in the post-mortem human brain: Insights from quantitative MRI and ICP-MS.
PG143 Iron concentration and longitudinal relaxation rate (R1) in the post-mortem human brain: Insights from quantitative MRI and ICP-MS.
Brain iron, primarily stored in ferritin and hemosiderin, plays crucial roles in cellular metabolism and accumulates with age (1,2), particularly in deep gray matter structures (3,4), such as the basal ganglia. While its influence on transverse relaxation times (T2, T2*) and quantitative susceptibility mapping (QSM) has been extensively investigated (5,6), its effect on the longitudinal relaxation rate (R1 = 1/T1) is still debated: while some studies have shown correlations with R1 in iron-rich regions (7–10), others failed to detect such relationships(11,12). This study explores the relationship between regional iron concentration and R1 in unfixed post-mortem human brains using quantitative MRI and chemical quantification via inductively coupled plasma mass spectrometry (ICP-MS).
Thirteen post-mortem human brains (mean age = 65.9 ± 10.2) were scanned in situ at room temperature at 3T using a 3D inversion recovery turbo spin echo sequence with TR = 8000 ms, TE = 8.5 ms, seven inversion times (TI = 100-3000 ms), and voxel size = 1×1×4 mm³. R1 relaxation rates were calculated via 3-parameter exponential fitting, and regions of interest (ROIs) were manually outlined to match tissue locations sampled for chemical iron quantification (Figure 1 shows a 1 cm-thick brain slice with an R1 map). After MRI, brains were extracted, immersion-fixed in formalin for 3-5 weeks, and bilateral specimens were collected from the globus pallidus, putamen, caudate nucleus, and from frontal, temporal, and occipital regions of both white matter and cortex. For statistical analysis, the globus pallidus, putamen, and caudate nucleus were grouped as basal ganglia (n=128); frontal, temporal, and occipital white matter as white matter (n=234); and analyzed cortical regions as cortex (n=71). Samples were freeze-dried, mineralized, and analyzed with ICP-MS (Agilent 7500ce), reporting iron in mg/kg of wet tissue. Statistical analyses included region-wise linear regressions, subject-level models, and linear mixed-effects models to account for inter-subject variability. Model assumptions (e.g., normality, homoscedasticity) were assessed. Mean iron concentration was highest in the basal ganglia (146 ± 51 mg/kg), followed by white matter (44 ± 12 mg/kg) and cortex (35 ± 12 mg/kg). Linear regression revealed a significant positive association between iron concentration and R1 in the basal ganglia (p < 0.001, R² = 0.15) and cortex (p = 0.011, R² = 0.09), but not in white matter (p = 0.051, R² = 0.02). Subject-level regressions showed consistent positive slopes in the basal ganglia and cortex. Mixed-effects models confirmed a significant relationship in both the basal ganglia and cortex, with inter-subject variability accounting for most of the explained variance (71% and 52%, respectively). To determine whether the association in the basal ganglia was driven by inter-regional iron differences, subregions analyses were conducted. The globus pallidus was the only subregion with a significant association with R1—unexpectedly with a negative slope. Our results confirm a rather low to non-existing correlation of iron concentration with the R1* relaxation rate. While a significant correlation was observed in pooled basal ganglia (Figure 2), the subregional analysis of the basal ganglia highlights the risk of aggregation bias (Figure 3), where the overall positive correlation may be driven by systematic differences in iron levels between subregions rather than reflecting a true continuous relationship. We hypothesize that other tissue properties such as myelin content (9,13,14), water content (6,13,15–17), or iron binding state (6,9,13) may additionally modulate the relationship beyond the rather low sensitivity for brain iron —a phenomenon found in QSM studies (14,18). Quantitative post-mortem MRI combined with ICP-MS confirmed a weak relationship between iron concentration and R1, mainly in pooled deep gray matter regions. However, this underscores the complexity of iron-related mechanisms on longitudinal relaxation and challenges the validity of R1 as a proxy for iron in the brain, compared with R2* or QSM.
Anna CAPPONI (Graz, Austria), Nikolaus KREBS, Walter GOESSLER, Eva SCHEURER, Kathrin YEN, Stefan ROPELE, Alessandra BERTOLDO, Christian LANGKAMMER
11:22 - 11:24
#47907 - PG144 Altered brain activation patterns associated to early-stage psychosis identified by working and verbal memory task-based fMRI.
PG144 Altered brain activation patterns associated to early-stage psychosis identified by working and verbal memory task-based fMRI.
Psychosis is characterized by delusions and hallucinations that alter the perception of reality. The first occurrence of such symptoms lasting over more than 7 days is termed first-episode psychosis (FEP) [1]. Psychotic disorders are highly heterogeneous, but depending on the presence of severe mood disturbances they are divided in affective (eg, mania with psychotic features or psychotic depression, both early stages of bipolar disorder) and non-affective (early stage of schizophrenia) psychoses [2].
Functional magnetic resonance imaging (fMRI) data can help understand alterations in brain mechanisms of both affective (A-FEP) and non-affective (NA-FEP) FEP. A recent review of advanced imaging studies in FEP reported hypoactivation in several regions during cognitive tasks, along with reduced DMN connectivity [1]. Further fMRI studies in FEP could help further characterize if functional brain disturbances previously reported in psychosis emerge during the early stages or during the course of the illness.
This study aims to examine functional brain differences between 1) healthy controls (HC) and patients with FEP; 2) A-FEP and NA-FEP individuals, using fMRI during a working memory task and a verbal memory task.
Eighty-two participants (aged 18-46) including 35 patients (13 A-FEP, 22 NA-FEP) and 47 HC were scanned in a 3T Siemens scanner. The protocol included T1-weighted image (TR = 2.3s, TE = 3ms, voxel size = 1x0.94x0.94 mm³) and two task-based functional MRI sequences (TR = 1.5s, TE = 37ms, voxel size = 2x2x2 mm³), a letter n-back working memory task and a verbal memory task.
During the n-back task, participants had to press a button when the letter shown to them matched the letter shown one (1-back) or two steps (2-back) back in a continuous sequence. The task consisted in eight blocks alternating between 1 and 2-back, each followed by a rest period marked by a cross. The verbal memory task included three periods of listening a list of incomprehensible words (rest), followed by four cycles of listening an understandable word list (encoding) and silent period of recalling words from the list (retention).
FMRI preprocessing included slice timing, motion correction, distortion correction (registration to the T1w image), spatial smoothing and frequency filtering. First-level activation maps were generated using nilearn, comparing task blocks. In the n-back task, 2-back and 1-back were compared with each other and with rest; for the verbal task, we assessed differences between retention and encoding, and between each phase and rest. FSL Randomise with threshold-free cluster enhancement was used to compare activation maps between HC and FEP, and between A-FEP and NA-FEP. Age, sex, study level and IQ were used as covariables and statistical significance at each voxel was set at p < 0.005. In the working memory task, reduced deactivation associated to the 2-back task was observed in the FEP in comparison to HC in medial frontal and right temporal regions (Figure 1). When comparing A-FEP and NA-FEP, greater activation was observed in the bilateral angular and supramarginal gyri in the A-FEP group, extending towards the left middle temporal gyrus (Figure 2). In the verbal memory task, decreased activation during memory retention was found in patients relative to HC in bilateral lateral occipital and right precentral areas (Figure 3). No differences were observed when comparing A-FEP and NA-FEP subjects. Task-based fMRI allowed to identify differences in the brain activation patterns of individuals with FEP with respect to HC both during working and verbal memory task. Individuals with FEP showed an increased activation during high cognitive working memory tasks in regions that follow a deactivation pattern, such as frontal regions typically associated to the DMN, a result similar with previous studies denoting DMN alterations [3]. Further alterations between FEP and HC groups were found in precentral regions related to verbal memory [4].
Analyses between FEP groups showed A-FEP having an increased activation in 2-back vs. rest in regions associated with a high activation pattern similar to the task-positive/dorsal attention network profile, which is known to be related to working memory. Literature supports task-positive network alterations in schizophrenia [5] and a stronger severity of cognitive impairment in schizophrenia [6]; therefore, a lower brain activation could be in line with this notion. These findings provide evidence of a specific pattern of brain activation in FEP patients already observable at early stages, that may relate to cognitive deficits. Functional MRI can be powerful to detect brain alterations in psychosis and identify specific patterns in N-FEP and NA-FEP. In this study, alterations were detected both by a working memory and a verbal memory task. Further studies will be performed to relate these brain activation patterns with cognitive deficits and to inform tailored early FEP interventions.
Alejandro HINOJOSA-MOSCOSO (Barcelona, Spain), M Florencia FORTE, Maria SERRA-NAVARRO, Derek CLOUGHER, Silvia AMORETTI, Eduard VIETA, Emma MUÑOZ-MORENO
11:24 - 11:26
#47654 - PG145 Quantitative MRI study of excised brain tissue in drug-resistant epilepsy patient: from in-vivo to ex-vivo.
PG145 Quantitative MRI study of excised brain tissue in drug-resistant epilepsy patient: from in-vivo to ex-vivo.
Quantitative MRI (qMRI) parameters have been used as in-vivo biomarkers to estimate tissue microstructure like myelin and iron[4]. To learn and validate the relation between qMRI and microstructure, ex-vivo high resolution qMRI maps have been compared to their histological counterpart[1,2]. However, the changes of qMR parameters from in-vivo to ex-vivo tissue and their dependence on image resolution (from mm to sub-mm) have not been fully characterised yet. This limits the translation of the the validated relation between qMRI parameters and histology to in-vivo applications. In the past, we explored the change of qMRI parameters from in-vivo to fixed ex-vivo for a freshly excised brain tissue section from drug-resistant temporal lobe epilepsy (dTLE) patients with close-to-clinical MR protocols[3,4]. Here we extend this study by (1) exploring a broader range of advanced qMR markers and (2) comparing with higher image resolution.
The study was acquired at the University Medical Center Hamburg-Eppendorf (ethics committee approval: protocol PV5600). The MR techniques used for all measurements were multi parametric mapping (MPM)[4], myelin water imaging (MWI)[5], magnitude-phase-based T2 mapping (MagPhT2)[6] and q-space trajectory imaging (QTI)[7]. Detailed information in Fig. 1A.
In-vivo subject (pre-surgery): A woman (56 y) diagnosed with drug-resistant temporal lobe epilepsy was measured with a 3T Prisma fit Siemens MR scanner and 64Ch head coil prior to undergoing hippocampal resection.
Ex-vivo specimen: A 16Ch wrist coil was used to measure the excised temporal pole (at room temperature) at three tissue stages: unfixed, fixed with a post-mortem interval of 45 min, and hydrated. The unfixed tissue was measured in glucose solution. Then, it was fixed with 4% paraformaldehyde (PFA) for 7 days (measured at the beginning and at the end of fixation). Later, the tissue was washed in phosphate buffered saline (PBS) solution for 2 days and measured in PBS + 0.1% NaN3. Finally, the tissue was scanned again using the same and high resolution protocols. Protocol details are in Fig. 1A.
Pre-processing and analysis: Several relaxometry and diffusion analyses (Fig. 1B) were performed for all MR measurements (Fig. 2A). Next, we affine-registered and resampled all the masked ex-vivo MR images (from unfixed to hydrated high resolution) to the in-vivo MR image (Fig. 2B). For the high resolution data and QTI, the parameter maps were re-aligned to in-vivo, but analysis was done in original space. According to Fig. 3, R1, R2, R2*, intra-axonal R2* (R2*-intra) and MWF increased after excision (in-vivo -> unfixed) and during fixation (fixed day 0 -> day 7), decreased after hydration (fixed day 7 -> hydrated), and preserved the cGM-WM contrast. Interestingly, myelin R2* (R2*-mye) behaves different (Fig. 3F): it increases after excision and when fixation started (day 0), but drops at the end of fixation (day 7) and even further during hydration. At increasing resolution (HydratedHighRes), R1, R2*-intra and R2*-mye increased barely, R2* decreased, and R2 and MWF remained stable.
In Figure 4, MD (Fig. 4C) dropped by more than half after excision, increased slightly during fixation, and decreased again during hydration. Notably, its cGM–WM contrast increased across all states, by increasing in cGM and decreasing for WM. uFA and MKA (Fig. 4B and F) showed a trend similar to R2*-mye (Fig. 3F) while the trend of the noisy MKI (Fig. 4D) was inconsistent. MKT (Fig. 4E), as a composite of MKI and MKA, followed the trends of its constituent parts. FA decreased in WM and increased in cGM after excision, then declined markedly in both during fixation. Hydration had no further impact. In high-resolution scans, GM–WM contrast was preserved, though absolute values shifted, presumably due to differences in protocol parameters. Our results confirm previous findings on how relaxometry, MWF, MD and FA changed across tissue stages (e.g., [21,23]); we also found a new common trend across tissue stages between MKa, uFA and R2*-mye. From these observations, we speculate that there is a common mechanism driving MWF and relaxometry on one side, and uFA, MKa and R2*-mye on the other side. Well-known candidates for the observed changes in relaxometry and diffusion parameters are reduced temperature, loss of perfusion, cellular apoptosis, cross-linking of proteins due to fixative, and reduced para-vascular space compared to in-vivo[20-23]. Finally, our high resolution results deviated from the low-resolution counterparts, some parameters more than others. Limiting factors of our study are the sample size (one), inaccuracies in registration between in-vivo and ex-vivo, and the use of in-vivo MR protocols for the ex-vivo specimen, which could result in parameter-estimation bias. Our comprehensive acquisition of qMR parameters revealed that transferring findings from ex-vivo to in-vivo MRI requires a thorough characterisation of their changes across tissue stages.
Francisco Javier FRITZ (Hamburg, Germany), Noémie Camille Rachel SURA, Nina LÜTHI, Laura BOGS, Laurin MORDHORST, Rüdiger STIRNBERG, José P. MARQUES, Filip SZCZEPANKIEWICZ, Jan Malte OESCHGER, Ora OHANA, Markus NILSSON, Evgeniya KIRILINA, Thomas SAUVIGNY, Siawoosh MOHAMMADI
11:26 - 11:28
#47630 - PG146 Study of the relationship between tumor metabolism modulators, IDO1, IDH and ChK-α, and the expression of the immune checkpoint, PD-L1, in glioblastoma models.
PG146 Study of the relationship between tumor metabolism modulators, IDO1, IDH and ChK-α, and the expression of the immune checkpoint, PD-L1, in glioblastoma models.
Immune checkpoint blockade-based immunotherapies (IMT) have demonstrated efficacy in some tumors such as melanoma (1), but have failed in others like glioblastoma (2), and we don't know the reason. A possible cause is aberrant tumor metabolism that allows tumors to create an immunosuppressive microenvironment (3). Previous studies showed a relationship between the immune checkpoint PD-L1 and key pieces of tumor metabolism (4). This work aims to investigate the existence of a relationship between aberrant tumor metabolism and acquired tumor immuneresistance.
We modulated the immune-checkpoint PD-L1 and three key metabolic enzymes, ChK-α, IDO1 and IDH1, to assess their effect on the lipid profile of various GBM models. We used the murine glioma cell line GL261 wild type (GL261wt) and IDH mutated (GL261mIDH) along with human glioblastoma cell lines (SF10602ML). Cells were seeded and treated for 48h with metabolic inhibitors, AGI-5198 and 1-MT, against mutated IDH and IDO1 respectively, or transfected with siRNAs against PD-L1 and ChK-α. Metabolic profiles were obtained through dual-phase metabolite extraction and subsequent 1H high-resolution NMR analysis (5). Spectra were acquired on a Bruker Avance Neo 11.7T NMR spectrometer. Integrals of the metabolites were determined and normalized to the TSP reference and the number of cells, from at least three experimental samples. Decreasing PD-L1 expression depicted increased levels of lipids involved in tumor progression, such as cholesterol or phosphatidylcholine in SF10602ML and GL261mIDH cells. Downregulating Chk- α increased the levels of total lipids in GL261-WT cells. Furthermore, the pharmacological inhibition of IDO1 with 1MT showed a significant increase of cholesterol, phosphatidylcholine and total lipids in GL261-WT cell line. On the other hand, the inactivation of mIDH1 with AGI-5198 reduced the total level of lipids in murine cell lines carrying the IDH1 mutation. Our results showed the existence of an interrelationship between PD-L1 expression and lipid metabolism in glioblastoma cells, highlighting the influence of the genetic profile on this interrelationship. This study demonstrates that PD-L1 has pro-oncogenic functions that go beyond its traditional role as an immunomodulator, influencing tumor metabolism. Metabolism also impact immunoresistence, as it has been demonstrated that lipids can reprogram T cells infiltrating the tumor mass towards immunosuppressive and anti-inflammatory phenotypes. Therefore, the increase in lipid levels upon PD-L1 downregulation could be utilized by tumor cells to regain resistance against the natural immune response. These results are highly relevant as they unveil a relationship between tumor metabolism and tumor acquired immuneresistance. The study of this relationship and the mechanisms that control it will allow us to understand why certain tumors do not respond to IMT, as well as rationally design new combinations of therapies seeking a synergistic effect. We hope to expand our research by working with new human cell lines carrying the IDH mutation, as well as by testing our results in glioblastoma in vivo models. Taking all this into account, we could demonstrate for the first time the existence of a relationship between tumor metabolism and PD-L1 expression in glioblastoma models.
Paula CARRETERO NAVARRO (Madrid, Spain), María José GUILLÉN GÓMEZ, Pilar LÓPEZ LARRUBIA, Jesús PACHECO TORRES
11:28 - 11:30
#47612 - PG147 Diffusion MRI derived white matter plasticity following mindfulness and inhibitory control training in adolescents.
PG147 Diffusion MRI derived white matter plasticity following mindfulness and inhibitory control training in adolescents.
Inhibitory control (IC), a core component of executive functions (EFs), plays a crucial role in cognitive and socio-emotional development [1]. Interventions such as cognitive training (CT) and mindfulness meditation (MM) have been shown to enhance IC [2-7]. IC relies on a widely distributed cortico-subcortical network, particularly involving prefrontal regions, which undergoes protracted maturation throughout adolescence [8-9]. Despite growing interest in IC training, little is known about how such interventions affect brain connectivity during this critical developmental window. Diffusion MRI (dMRI), which enables in vivo assessment of white matter (WM) microstructure, is well suited for studying structural connectivity. In this study, we investigated the effects of five weeks of computerized MM, CT, or active control (AC) training on WM microstructure in adolescents.
91 healthy adolescents (16–17 y.o., 55 females) were recruited. Participants were randomly assigned to MM, CT, or AC groups. EFs were assessed with cognitive and emotional Stroop task, Simon task, Delay Gratification Tasks, Trail Making Test, the dot Task, the Stop Signal Task, and the N-back task. Diffusion MRI data (single shot spin-echo echo-planar imaging, 30 directions, TR = 11s, TE = 0.0867 s, flip angle = 90, b-value = 1500 s/mm2, slice thickness = 2 mm, voxel size = 2.0 × 2.0 × 2.0 mm) were collected pre- and post-intervention. dMRI data were preprocessed with FSL using standard parameters [10]. Individual maps of fractional anisotropy (FA), mean diffusivity (MD), radial diffusivity (RD), and generalized fractional anisotropy (GFA) were then quality checked and analyzed using a longitudinal adaptation of the standard Tract-Based Spatial Statistics (TBSS) approach [11]. Maps comparisons along with correlations between cognitive changes and dMRI metrics changes were assessed using voxelwise permutation testing (n = 5000, TFCE-corrected, p < 0.05) and anatomical labeling based on the JHU WM atlas [12-14]. No significant changes in WM microstructure were observed within any of the training groups when analyzed independently. However, we found significant differences when comparing WM changes across groups, as well an association between WM alterations and EF changes across the different training conditions. Compared to CT, MM exhibited greater increases in FA and decreases in RD, particularly within the forceps minor, anterior thalamic radiation, and inferior fronto-occipital fasciculus. In addition, we found significant differences between conditions in main WM tracts associated with cognitive control, such as the ILF, SLF, IFOF, cingulum and anterior thalamic radiation in the following tasks: N-back, cognitive Stroop, SST and Simon. These findings provide evidence for short-term WM microstructure plasticity in adolescents and suggest that different interventions modulate distinct neuroanatomical pathways. The WM pathways affected by the interventions are known to be critical for executive functions and interhemispheric integration. Of note, MM appeared to induce more robust enhancements in WM microstructure. The emerging pattern of training-specific plasticity highlights the importance of tailoring interventions based on targeted specific cognitive domain. Our results provide diffusion MRI evidence that both MM and CT can induce short-term WM plasticity during adolescence, with distinct effects across major white matter pathways. These findings enhance our understanding of how different interventions can shape brain development and inform the design of targeted, evidence-based strategies to promote mental health and cognitive functioning in youth. The use of dMRI biomarkers enables the detection of subtle, training-induced changes in WM microstructure. Ongoing analyses in children (to be presented at the meeting) will further investigate potential age-related differences in WM plasticity and test the hypothesis that training may accelerate WM maturation.
Belen AZOFRA-MACARRON (Paris), Lorna LE STANC, François RAMON, Gabriela REZENDE, Iris MENU, Cloélia TISSIER, Emilie SALVIA, Julie VIDAL, Marine MOYON, Lisa DELALANDE, François ORLIAC, Nicolas POIREL, Catherine OPPENHEIM, Olivier HOUDÉ, Grégoire BORST, Arnaud CACHIA
11:30 - 11:32
#46640 - PG148 Sleepy QSM: Study to assess the effect of sleep deprivation on brain homeostasis with 7T QSM and qT1.
PG148 Sleepy QSM: Study to assess the effect of sleep deprivation on brain homeostasis with 7T QSM and qT1.
Acute sleep deprivation (>24 h) consistently slows reaction times and increases lapses on the Psychomotor Vigilance Task (PVT), indicating impaired sustained attention.[1] During normal sleep, glymphatic exchange clears neuro-metabolic waste, whereas MRI T1-mapping shows this clearance is attenuated when wakefulness is prolonged.[2][3] Quantitative susceptibility mapping (QSM) has revealed sleep-related microstructural changes in brainstem and subcortical nuclei in disorders such as REM sleep behaviour disorder and obstructive sleep apnoea, underscoring its sensitivity to iron and perfusion shifts that accompany disrupted sleep physiology.[4][5]
This study investigates the effects of 24h of sleep deprivation on regional QSM and T1 alterations to concurrent cognitive decline using 7T MRI. We therefore combined 7T QSM/T1 mapping with repeated PVT assessments in healthy adults to test reversible, region-specific changes that track the magnitude of cognitive slowing.
We enrolled 30 healthy subjects (age 20-38) who underwent an 7T MRI at baseline (day 0), after sleep deprivation of 24h (day 1) and post recovery (day 4). Participants were supervised by medical staff to confirm that the participants were awake for all 24h. The impact of sleep on cognitive performance was measured using reaction time assessments from NASA PVT (300s duration each) at 0h, 12h, 24h and 96h, extracting mean latency, the slowest 10 % and fastest 10 % of responses as performance endpoints. 7T QSM was computed from complex images acquired with an ASPIRE-based GRE sequence (0.7x0.7x0.7mm^3, TE=5, 10, 15, 20ms, TR=25ms) [6] and reconstructed with QSMbox [7].
Per time point, an additionally acquired MP2RAGE (0.7x0.7x0.7mm^3) was used to generate a subject-based whole-brain segmentation via FastSurferCNN [8]. The created segmentation was transformed to the QSM data using FSL FLIRT [9]. The ROIs were used to calculate the average mean +/- stdev QSM values. In addition, the average mean +/- stdev was also determined from the T1 maps of the MP2RAGE sequence.
Spearman’s rank correlation coefficient (r) was computed to assess monotonic associations between changes in reaction times (ΔRT) and ΔQSM or ΔT1 for two time intervals (day 0 vs. 1 and day 1 vs. 4). All pre-processing steps and the analysis were performed with Python. Sleep deprivation led to a transient increase in mean reaction times, including both the slowest and fastest 10% responses at 24 h, with full recovery by 96 h (all p > 0.05; Fig. 1). Reaction time variability decreased progressively, consistent with a training effect.
Quantitative susceptibility mapping (QSM) revealed a significant negative correlation between ΔQSM (Fig. 2) in the ventral diencephalon and slowing in the slowest 10% of responses (r = –0.40, p = 0.029), indicating regional susceptibility changes associated with cognitive impairment. Trends toward significance were also observed in the thalamus proper (r = –0.30, p = 0.107) and cerebral white matter (r = –0.31, p = 0.095). No significant associations were found with mean or fastest 10% reaction time changes.
T1 relaxometry showed the strongest effects in the lateral ventricles (Fig. 3), where T1 prolongation correlated significantly with both mean reaction time (r = –0.48, p = 0.007) and the slowest 10% (r = –0.37, p = 0.041). A near-significant trend was seen in the ventral diencephalon (r = –0.36, p = 0.050). No significant correlations were found for the fastest 10% reaction time decile. QSM-based susceptibility changes in the ventral diencephalon were significantly associated with cognitive slowing during sleep deprivation, implicating an involvement of deep gray matter in transient, sleep deprivation-induced, transient functional impairment. These alterations may reflect regional changes in iron content, perfusion or glymphatic flow. Supporting trends in thalamic and white matter regions suggest a more widespread but reversible vulnerability of subcortical structures to sleep-related stress, particularly affecting attentional maintenance as reflected by the slowest response decile.
T1 prolongation in the lateral ventricles showed robust associations with slower reaction times, highlighting ventricular fluid shifts or altered CSF-interstitial exchange as potential contributors to cognitive decline. The near-significant T1 findings in the ventral diencephalon parallel the QSM results, underscoring this region’s relevance in sleep-related brain physiology. The lack of associations with the fastest reaction times suggests these MRI metrics are more sensitive to processes underlying sustained attention and fatigue. Sleep deprivation induces region-specific, reversible MRI changes in susceptibility and T1 relaxation that correlate with cognitive slowing, particularly in the ventral diencephalon and periventricular regions. These findings support a role of altered glymphatic function and subcortical vulnerability in mediating the cognitive effects of sleep loss.
Eric EINSPÄNNER, Hendrik MATTERN (, ), Erelle FUCHS, Sebastian MÜLLER, Eya KHADHRAOUI, Daniel BEHME
11:32 - 11:34
#47857 - PG149 fMRI analysis of cerebrospinal fluid flow during slow paced breathing compared to free breathing.
PG149 fMRI analysis of cerebrospinal fluid flow during slow paced breathing compared to free breathing.
Cerebral spinal fluid (CSF) pulsations are associated with brain waste clearance and may be driven by breathing, among other factors such as heart rate, vasomotion, autonomic function or neuronal activity [1,2]. In an impactful study, Fultz et al. showed that fMRI can be used to assess the pulsatile inflow of CSF into the fourth ventricle and its coupling with the low-frequency global grey matter BOLD signal (gBOLD), presumably reflecting a mechanism whereby reductions in total cerebral blood volume (CBV) lead to the inflow of CSF into the brain to preserve the constant volume of fluids in the head [3]. Using this approach, recent studies showed that, besides sleep as in the original paper, breathing can modulate CSF flow and the CSF-gBOLD coupling, including brief deep breaths as well as paced breathing and breath-holding [4,5]. Such breathing modulations seem to amplify CSF flow, potentially as a result of the associated intrathoracic pressure and blood CO2 changes [4,5]. Here, we aim to further investigate how autonomic modulation through a slow paced breathing (SPB) task affects CSF flow and the CSF-gBOLD coupling, compared to free breathing rest (FBR).
fMRI data were collected from 15 healthy women (30.9 ± 6.8 years) during the two phases of their menstrual cycles (before menses and post-ovulation), during: SPB (2 min at 0.1 Hz, preceded and followed by 1 min of FBR, with visual instructions), and FBR (7min eyes open) in the awake state. fMRI data was collected in a 3T Siemens Vida scanner with a 64-channel RF coil using 2D-EPI (TR/TE=1260/30ms, in-plane GRAPPA-2, SMS-3, 60 slices, 2.2mm iso resolution). Moreover, respiratory signals (integrated Siemens Biomatrix sensors) were continuously recorded during the scans. One phase of one subject was excluded due to a technical issue. Since no differences were found between the two phases of the menstrual cycle, the two sessions were pooled for subsequent analysis. fMRI data processing included (code [6]) motion and distortion correction, brain extraction, motion outliers detection, temporal high pass filter (0.01 Hz), spatial smoothing (3.3 mm - FBR, 3.5 mm - SPB), and registration. A Butterworth bandpass filter 0.01-0.2 Hz using a zero delay fourth order was also applied. gBOLD was obtained by averaging the BOLD signal across the cortical grey matter (automatic segmentation using FSL’s FAST). The CSF signals were extracted from the fourth ventricle (semi-manually defined). Lagged cross-correlations were computed using Pearson’s correlation coefficient to assess signal coupling between gBOLD (and its negative derivative) and CSF signals. Similar analyses were conducted between respiratory and gBOLD signals, as well as between respiratory and CSF signals. Large modulations in the CSF and gBOLD signals occur alongside corresponding large changes in respiration during SPB, while smaller amplitude modulations are observed during FBR, as seen in Fig. 1. Fig. 2 shows the cross-correlation curves for both tasks. Consistently with the literature, during FBR, the first peak in the gBOLD–CSF cross-correlation is positive, followed by a negative peak. In contrast, this pattern is reversed during SPB, with an initial positive peak followed by a negative one. Fig.3 depicts the distributions across subjects of the cross-correlation values of the first and second peaks, ordered by lag time, as well as their lags, during FBR and SPB. Higher gBOLD-CSF correlation values were obtained during SPB relative to FBR. Despite differences in lag, clear anticorrelation of the gBOLD and CSF signals was observed during both conditions, consistent with previous studies. As shown in Fig.4, compared to FBR, SPB exhibits a stronger coupling between respiratory signals and CSF and gBOLD signals. The gBOLD–CSF coupling described in the previous human studies is also present in our dataset during rest [1,2]. However, the breathing manipulation increased the coupling strength while shifting its lag. SPB shows a clearer, more symmetrical, and stronger correlation centred around 0 s between the gBOLD derivative and CSF signals. This means that rapid changes in gBOLD tend to occur at the same time as changes in CSF, suggesting a tighter temporal link between gBOLD-CSF dynamics during SPB. SPB enhances both positive and negative cross-correlation peaks compared to FBR, suggesting stronger gBOLD–CSF coupling. Overall, our results suggest that autonomic activity associated with this slow paced breathing task at 0.1 Hz, which engages the parasympathetic system, may contribute to CSF pulsations, further supporting the findings of the only previous study of paced breathing [4]. Future studies should clarify the timing differences observed in the gBOLD-CSF coupling during such a breathing task when compared to rest with free breathing. We observed that SPB significantly enhanced gBOLD–CSF coupling relative to FBR, suggesting a link between respiratory-driven autonomic modulation and CSF dynamics.
Maria DIAS (Lisboa, Portugal), Inês ESTEVES, Frederico SANTIAGO, Sara MONTEIRO, Ana FOUTO, Amparo RUIZ-TAGLE, Gina CAETANO, Patrícia FIGUEIREDO
11:34 - 11:36
#47905 - PG150 In vivo test-retest study of liver stiffness in male and female mice using two motion encoding MR Elastography methods.
PG150 In vivo test-retest study of liver stiffness in male and female mice using two motion encoding MR Elastography methods.
Magnetic Resonance Elastography (MRE) is a non-invasive technique used to assess tissue stiffness, particularly in the context of liver fibrosis [1]. We have developed an MRE approach based on optimal control (OC) theory, enabling motion encoding at short echo times (TE) and improving signal-to-noise ratio (SNR). To support future clinical translation, this method must first be validated in animal such as murine models of liver diseases. In this work, we perform in vivo liver MRE acquisitions in mice we compare the performance of RARE-based MRE sequences using the classical motion encoding gradient method (MEG-RARE) and our optimized OC-based approach (OC-RARE) [2–4] through a test-retest protocol. We also examine stiffness measurements (shear storage modulus G′) and SNR variability between male and female mice.
Six healthy mice (3 females, 3 males) were examined in accordance with ethical standards. The average weight of the female mice was 25 g (born in November 2024), while the male mice weighed an average of 30 g (born in October 2024). Anesthesia was maintained using 2% isoflurane, with continuous monitoring of respiratory rate and body temperature. MRE acquisition were performed on two different days (D1 and D15) at 300 Hz using two sequences: the classical motion encoding gradient method (MEG-RARE) and the optimal control-based method (OC-RARE). In the latter, RF pulses generated via an optimal control algorithm simultaneously ensured slice selection and motion encoding. Acquisition parameters are detailed in Figure 1. Mechanical wave motion was encoded along the slice direction using two opposite wave polarities, enabling phase subtraction to reduce phase noise. Phase images were then processed through phase unwrapping, temporal Fourier transform, and spatial filtering, before elastogram reconstruction using the AIDE algorithm [5]. G′ values were measured in liver regions where wave displacement along the slice direction exceeded 5 µm, corresponding to a minimum phase shift of approximately 0.93 radians in the MEG-RARE sequence. Variations in G′ were analyzed across sequences, sexes and acquisition timepoints. To assess measurement precision, a global coefficient of variation (CV) was calculated for each method using all repeated G′ measurements across animals, defined as:
CV (%) = (SDG’ / mean G’) × 100, where SDG’ is the standard deviation of repeated measurements.
Test-retest reproducibility was evaluated using the relative variation ΔG′ between Day 1 and Day 15, defined as:
ΔG′ (%) = [(G′_D1 − G′_D15) / ((G′_D1 + G′_D15)/2)] × 100. Figure 2 and 3 illustrate representative magnitude images, wave images, and elastograms obtained from one female (Fig. 2) and one male mouse (Fig. 3) at Day 1. Figure 1 summarizes the mean G′ values, standard deviations, global CV, ΔG′ and signal-to-noise ratios (SNR) for each mouse and acquisition method. Both MEG-RARE and OC-RARE showed good test–retest reproducibility, with slightly lower ΔG′ and CV values observed in the OC-RARE method. OC-RARE also provided a higher signal-to-noise ratio (SNR ≈70) compared to MEG-RARE (SNR ≈40). It can be observed that G′ values were moderately higher in males than in females, as seen in Figure 4 but that for male mice, results between D1 (five months old) and D15 are equivalent across both methods whereas for female mice, G’ values increase between D1 (four months old) and D15. The results show that the OC-RARE method, based on motion encoding via optimal control, provides liver stiffness measurements comparable to the classical MEG-RARE method while significantly improving the signal-to-noise ratio. The results demonstrate stable G′ measurements across two timepoints, supported by low global CV and ΔG′ values. The SNR improvement is mainly related to shorter echo times (TE) enabled by motion encoding without motion encoding gradients. Finally, the optimal control approach appears promising for improving MR elastography acquisition quality. Our results show that for female mice which are four months old, liver stiffness changes whereas usually, it is taken for granted that as from two months age, female mice are adults implying stable liver stiffnesses. The OC-RARE method enables reliable magnetic resonance elastography acquisition, providing liver stiffness measurements comparable to those obtained with the conventional MEG-RARE approach. It demonstrates good measurement precision and satisfactory test-retest reproducibility, while offering higher signal-to-noise ratios. By improving overall acquisition quality without compromising measurement accuracy, the optimal control-based encoding strategy shows particular promise for studying tissues with short T2 relaxation times, such as iron-overloaded liver. Further studies in larger cohorts and pathological models are needed to fully validate its potential in preclinical settings. The study showed the importance of using both sexes and taking their ages into account.
Tiffany BAKIR AGERON (Lyon), Kevin TSE VE KOON, Pilar SANGO-SOLANAS, Olivier BEUF
|
Salle 120 |
|
"Friday 10 October"
Added to your list of favorites
Deleted from your list of favorites
E22
11:00 - 12:30
MS3 - Sensing myelin with MRI
Different perspectives and the actual needs
|
Salle 76 |
|
"Friday 10 October"
Added to your list of favorites
Deleted from your list of favorites
G22
11:00 - 12:30
Poster 3
FT1 - RF hardware | FT1 - Hardware beyond RF | FT1 - Cross-modality technologies | FT3 - Phantoms & Simulations
11:00 - 12:30
#47888 - PG319 Design and demonstration of a tunable metasurface for improving abdominal imaging at 3 T.
PG319 Design and demonstration of a tunable metasurface for improving abdominal imaging at 3 T.
At 3 T MRI, the radiofrequency (RF) wavelength becomes comparable to body dimensions, causing dielectric artifacts and dark regions in images. Recent work suggests using metamaterials [1,2] and metasurfaces (MSs) [3] to shim the transmit field passively. Placed on the body, these structures localize the RF field within the ROI, increasing |B₁⁺| amplitude and mitigating dielectric artifacts. Here, we propose an MS comprising six concentric split-ring resonators (SRRs). This configuration offers two key advantages: easier tuning using four capacitors per SRR and adjustable RF field distribution through frequency detuning (Δf) between SRRs. By varying Δf between adjacent rings, the system achieves tailored B₁⁺ field distributions optimized for specific anatomical ROIs.
Numerical simulations were performed in CST Studio Suite 2022 using a frequency domain solver. The MS consisted of six square concentric SRRs with side lengths of 46, 90, 134, 178, 222, and 266 mm, respectively. Each SRR was made of 2 mm-wide copper strips and included four gaps loaded with variable capacitors (Fig.1a). The resonant frequency of the entire device was tuned to 123 MHz. A detuning approach was applied by selecting the capacitance values such that each inner SRR had a first resonant mode at a higher frequency than its neighboring outer ring. Several configurations with different frequency gaps (Δf) between adjacent SRRs were studied (Fig.1b).
To estimate the |B₁⁺| and the SAR distributions, we performed numerical simulations using a voxelized human model ‘Duke’ (Sim4Life) with realistic tissue properties. The transmit RF coil was a 16-leg shielded high-pass whole-body birdcage coil with a 700 mm bore diameter (Fig.1c). The |B₁⁺| and SAR distributions were normalized to 1W of the accepted power.
The coefficient of variation (Cv) was used to assess |B₁⁺| inhomogeneity. It was calculated as the standard deviation divided by the mean |B₁⁺| in the ROI, expressed as a percentage. The local maximum SAR (SARav.10g) was calculated as averaged over 10g of tissue. SAR efficiency was determined as the |B₁⁺|/√SARav.10g.
Experimental validation was carried out on a 3 T Siemens Magnetom Trio system using a birdcage coil for transmission and a matrix surface coil for reception. The MS configuration with Δf = 7 MHz was selected for experimental studies based on its optimal SAR efficiency in simulations. The HASTE pulse sequence (TE/TR = 61/2000 ms; FA = 180°; FOV = 400×400 mm²; acquisition matrix = 384×384; slice thickness = 5 mm) was used to acquire abdominal MR images of a healthy volunteer with and without the MS. The SNR maps were calculated by dividing the mean signal intensity in the liver region by the standard deviation of the noise estimated in signal-free background areas. Numerical simulations showed that the MS increased the |B₁⁺| amplitude in the liver from 0.15 to 0.19–0.20 µT (~35% gain). Cv of the B₁⁺ field in the liver was 14.7% in the reference case and changed to 21%, 17.5%, and 19% for Δf = 0, 7, and 20 MHz, respectively.
SAR maps (Fig. 3) showed localized hotspots near the liver in all MS configurations. The SARav.10g maximum increased from 0.14 W/kg (reference case) to 0.23, 0.20, and 0.21 W/kg for Δf=0, 7, and 20 MHz, respectively. However, the reference case showed SAR efficiency of 0.40 μT/√W/kg, while the MS configurations demonstrated SAR efficiencies of 0.40, 0.45, and 0.43 μT/√W/kg for Δf=0, 7, and 20 MHz, respectively.
Experimental studies of liver MRI demonstrated that using the MS increased the average SNR in the ROI by 46% compared to the case without the device (Fig. 4). Numerical simulations revealed that all considered MS configurations effectively reshaped the B₁⁺ field of the birdcage coil, producing an increase in B₁⁺ field amplitude within the ROI. This redistribution successfully compensated for the B₁⁺ field minimum present without the MS. Although each configuration increased the local SARav.10g, the concurrent enhancement of the B₁⁺ field amplitude led to improved SAR efficiency. The configuration with Δf = 7 MHz demonstrated the highest SAR efficiency of 0.45 μT/√W/kg, making it the optimal choice for experimental implementation.
Experimental MRI studies with a healthy volunteer demonstrated that the proposed MS enhanced signal intensity in the ROI.
The current study focused on optimizing the MS's configuration specifically for liver imaging. However, the proposed tunable MS offers broader applicability due to its flexible adaptability to different ROIs. By modifying capacitor values, the spatial distribution of the RF magnetic field can be precisely controlled, allowing customization for different anatomical regions. The tunability and the structure's relative simplicity make the design versatile and practical for clinical implementation.
This study was supported by the state assignment No. FSER-2025-0018 within the framework of the national project “Science and Universities,” Russian Federation.
Leila SHARIPOVA (Saint Petersburg, Russia), Alena SHCHELOKOVA, Viktor PUCHNIN
11:00 - 12:30
#47610 - PG320 Multi-channel receiver array with wireless connection to the patient table.
PG320 Multi-channel receiver array with wireless connection to the patient table.
Although MRI is one of the most valuable and versatile imaging modalities, it also has considerably higher operational costs in comparison to for example CT or ultrasound. The relatively long duration of MRI investigation and the need for highly trained staff are the dominant contributors to the operational costs of a MRI system. Reducing setup time and simplifying the setup procedure may help to reduce MRI costs. One of the ways to simplify the patient setup is by removing the need to connect RF coils. Approaches that use integrated circuits to digitize the MRI signal on the coil and use conventional wireless signal transfer have been proposed, but come with i.e. lack of required bandwidth, complex circuitry and full redesign of the MRI receiver chain.
A simpler approach is to use the RF body coil that through mutual inductance with a local receiver can obtain the signals from the local resonant coil via the receiver connected to the RF body coil [1,2].
However, this hinders the use of wireless local receiver arrays. Inductive matching has been proposed and implemented for more than 30 years [3,4] in fixed mechanics between the receiver coil and the matching coil. Last year we demonstrated that such mechanics can be made variable while maintaining good matching conditions between the receiver coil and the inductively coupled matching loop [5].
In this work, we investigated if an array of stretched coils that have an inductive coupling with loops made inside the bed can be used as a simple approach for wireless RF coil array operation. More specifically, the feasibility of wireless reception using a 4-channel wireless coil with 4 independent receiver loops was investigated.
A 4-channel wireless coil (6x47cm each channel), (Fig.1.A) was tuned and matched at 64MHz for a 1.5T system (Ingenia, Philips, Best, The Netherlands) when it was loaded with a body phantom (σ=0.55S/m, ε=74 at 100MHz). Decoupling via overlapping was implemented. A passive detuning (BAV99W, Eindhoven, The Netherlands) was applied (Fig.1.B) to decouple each channel from the transmit coil during RF-transmit.
Two pairs of 5 cm diameter receiver loop coils were designed (Fig.1.C). Each one was tuned and matched at 64MHz when it was placed 2 cm away from the loaded wireless coil (mimicking the patient table). A detuning circuit was applied (Fig 1.D). Overlapping was implemented to reduce coupling between loop coils.
The S-parameters of the receiver loop coils and the 4-channel wireless coil, including efficiency and decoupling, were measured as in Setup1 (Fig.2.A) and Setup2 (Fig.2.B).
Phantom Imaging
The wireless coil was placed around the phantom. Receiver loop coils were placed 2 cm above the wireless coil and only loop coils were connected to the interface box (Fig.2.C). 2D-Dynamic-GRE sequence was acquired. SNR maps per channel and noise correlation matrix were calculated. All VNA measurements are given in Fig.3. S21 of Setup1 when the receiver loop coil was placed 2 cm above the wireless coil was -27 dB for all channels (Fig.3.A). The decoupling between channels of the wireless coil of Setup2 was at least -13 dB (Fig.3.B). S11 of each receiver loop coil was less than -14 dB, the decoupling between receiver loop coils was at least -14 dB (Fig.3.C).
SNR maps obtained from each channel of the wireless coil, particularly in the coronal plane, show signal intensity peaks at the edges corresponding to the sensitivity profiles of both receiver loop coils and wireless coils (Fig.4.A). The noise correlation matrix shows low noise correlation between the channels (Fig.4.B), resulting in largely independent behavior of the receiver loop coils, which is also in agreement with bench measurements. This work presents an implementation of wireless MR signal reception via inductive coupling for a 4-channel wireless coil. The bench results of Setup1 and Setup2 show the applicability of the wireless array coil design when all channels are independent. SNR maps and noise correlation matrix show the possibility of independent reception of each channel of a 4-channel wireless coil in phantom studies. It provides a perspective for further steps in the design of a wireless coil array and its accompanying receiver loop coil array that would accelerate and simplify studies of complex regions of the human body. In conclusion, this study has shown that all 4 wireless channels can operate independently, allowing true multi-channel reception with high sensitivity to be achieved over a wide area. The results presented may provide an impetus for the development of wearable coil array technology.
Nikolai LISACHENKO (Utrecht, The Netherlands), Alexander RAAIJMAKERS, Dennis KLOMP, Busra KAHRAMAN-AGIR
11:00 - 12:30
#47082 - PG321 An 8-channel transceiver array made of high-impedance dipoles for 11.7T brain MRI.
PG321 An 8-channel transceiver array made of high-impedance dipoles for 11.7T brain MRI.
The first in-vivo human brain images at 11.7 T were recently shared to the community [1]. Such a high magnetic field comes with several challenges, in particular regarding B0-related artifacts in fast imaging sequences like EPI. In this context, a 27-cm internal diameter multi-coil-array (SCOTCH) was developed [2]. To avoid cross-interactions, a shield must be placed between this device and the RF coil. In this abstract, we propose an 8-channel transceiver array designed to fit inside SCOTCH and made of high-impedance dipoles (HIDs) paired with transmission line baluns, both avoiding the cumbersome use of lumped components.
Based on the same principle as high-impedance loops [3,4], we introduce the HID as a transmission line structure (here as a buried microstrip) with two gaps on its outer conductor making it resonant at the Larmor frequency (499.4 MHz). One can fine tune the HID either by changing the gaps’ positions or their widths. In this simulation, a 6.15 dielectric constant material (Rogers RO4360G) was used for the HID microstrip, making it 26.2 cm long. At 499.4 MHz, the HID is tuned so that the imaginary part of its impedance is equal to 0, and its real part is about 800 Ohms, providing the high-impedance property (Fig. 1A). A transmission line balun (“Marchand balun” [5]) transforms the 800 Ohms to 50 Ohms both required for maximum power transfer at Tx, and for optimal noise matching at Rx (Fig. 1B). A transmit-receive (T/R) switch (Fig. 1C) is integrated into the stripline structure, using three diodes. In the transmit mode, the diodes are biased and protect the preamplifier input (34 dB isolation). In the receive mode, a close to λ/2 long transmission line transforms the low-input impedance of the preamplifier (1.5 Ohms) to a high impedance at the balun’s input, then again transformed into a low-input impedance at the HID port, ensuring preamplifier decoupling between the elements. The stripline structure embedding the balun and the T/R switch is duplicated 16 times with a 20° azimuthal shift. Eight HIDs are connected to every other balun structure; the leftover structures merely close the RF shield and ground plane for the coil. At the front, a copper foil is added to electrically close the shield while saving some space to include a mirror for visual stimulation.
Using HFSS (Ansys, PA, USA), the coil was simulated with two homemade multilayers head and shoulders models (Fig. 2). The elements were geometrically fine-tuned on the female model, and no adjustment was done after changing the model to the male one. For completeness, two different HFSS simulations were performed: one at the Tx port, and one at the Rx port (Fig. 1B), both taking into account the parasitic diode effects: a 0.4 pF capacitance in the receive mode, and a 0.4 Ohm resistance in the transmit mode. In each case, the ports were driven with 1W input power. The H-field maps were exported with a 5-mm voxel resolution and transformed to B1+ and B1- profiles. In the receive mode, the S matrix was exported to compute the SNR [6]. Virtual Observation Points (VOPs) were computed to run SAR-constrained kT-points [7] pulse design for different flip angles (maximum average power per channel = 6 W, maximum 10g-SAR limit = 20 W/kg). In the transmit mode, all elements are matched to lower than -10 dB, ensuring no excessive reflection (Fig. 3A); in the receive mode a low noise correlation is measured in each case and can be attributed to the distance between dipoles (Fig. 3B). The sum-of-magnitudes and SNR maps both exhibit a fairly good homogeneity, except at the top of the brain, which lacks some signal. This signal dropout in this region is more important for the male model and translates into some B1+ shimming difficulties using pulse design (Fig. 4). A perpendicular-to-B0 shield electrically connected to the baluns’ ground plane at the back of the coil should help to retrieve some signal in the upper brain regions. Even though not yet available at our 11.7T system, a 16-channel transceiver array would certainly allow to further mitigate B1+ inhomogeneities. In that case, a z-segmented array could also be beneficial to give more freedom to the pulse design optimization. A complete simulation model of an innovative transceiver array at 11.7T was presented. The newly introduced high-impedance dipole paired with a transmission line balun allowed to suppress bulky lumped components. This transceiver array will be later on paired with a tight-fitting cap receive array to maximize SNR at the periphery of the brain [4].
Paul-François GAPAIS (Paris), Michel LUONG, Alexis AMADON
11:00 - 12:30
#47930 - PG322 Towards $50 NMR: commercial sdr and preamplifiers for cost-effective mr signal reception with active rx/tx switching.
PG322 Towards $50 NMR: commercial sdr and preamplifiers for cost-effective mr signal reception with active rx/tx switching.
Magnetic resonance spectroscopy (MRS) and tomography (MRT) are valuable techniques for analyzing chemical and biological samples but are often only available to specialized laboratories due to their high cost and complexity [1,2]. To overcome these barriers, it is needed need to develop more cost-effective and accessible NMR technologies [3]. One approach is the use of Software-Defined Radio (SDR), since it offers a promising solution to increase the flexibility and efficiency of NMR analyses [4]. It was also shown that combining pulsed and continuous wave NMR with active RX/TX-switching can improve the signal-to-noise ratio [5].
To develop a more cost-effective solution and to reduce complexity we present a MR signal reception path based on commercial (SDR) sticks, commercial preamplifier, and a custom-built active transceiver (RX/TX) switch tested with a low-field MR system.
For the measurements, an MR signal reception path consisting of a commercial SDR USB stick (NESDR SMArt v5 SDR, Nooelec Inc., Wheateld NY, USA), commercial broadband preamplifier (827becxfh, Walfront, China) and a custom-built active RX/TX switch was developed. The coil, magnet, and transmit pulse generator from a low-field benchtop MR system (MagSpec 0.57 T & Drive L, PureDevices GmbH, Rimpar, Germany) were taken for the experiments. The TX pulse is transmitted through the RX/TX switch into the coil, while the RX signal is routed through the RX/TX switch, passed through the preamplifier, and then directed into the SDR stick. The SDR stick is controlled using GNU-Radio [6], while the TX signal generation and RX/TX switching are managed via benchtop MR in MATLAB software (MathWorks Inc., Natick, MA, USA). The measurement setup is shown in Figure 1.
The RX/TX switching is achieved using two PIN diodes. In TX mode, a direct current is applied to PIN diodes D1 and D2, forming a parallel LC circuit with the low-pass pi-circuit components (C1, L, C2). D2 short-circuits C2, preventing strong TX signals from reaching the preamplifier. In RX mode, both diodes are reverse biased, suppressing noise from the power amplifier. Figure 2 shows a schematic of the RX/TX switch. The TX/RX-switch was analyzed with a vector network analyzer (ZNL3, Rohde & Schwarz GmbH, Munich, Germany).
To evaluate the MR signal reception capabilities of the SDR stick, a non-selective multi-echo T2 sequence was employed with following parameters: Rectangular Pulses, TE: 4 ms, 38 echoes. During the sequence, raw data containing both (suppressed) TX and RX signals were acquired using GNU-Radio. This raw data was subsequently analyzed in MATLAB, and the T2 relaxation curves were plotted. The experiment was conducted using tap water, a 0.25 mmol/mL contrast agent solution (gadofosveset) in water, and an oil phantom (PureDevices). Our signal reception setup successfully captures signals, confirming its basic functionality, although signal quality is affected by the lack of electromagnetic shielding, which introduces noise.
The impedance of the TX/RX switch matches 50 Ω while in RX mode / PIN diode switched off (S11: < - 60 dB) and low transmission loss (S21: - 0.28 dB) and is mismatched while in TX mode / PIN diode switched on (S11: - 0.58 dB) and high transmission loss (S21: - 31.15 dB). S11 was measured from coil and preamplifier port, S21 from preamplifier to coil port. Both return and transmission loss and the corresponding Smith chart are shown on Figure 3. The T2 values amount as, 930.16 ± 38.21 ms for tap water, 96.95 ± 1.03 ms for the CA solution, and 144.58 ± 17.12 ms for the oil phantom. These values agree with values measured solely with low-field MR system (tap water: 915.0 ms, CA solution: 97.67 ms, Oil: 141.5 ms) and literature values at 0.5 Tesla, which range for water (500-900 ms), CA solutions (40-100 ms), and oil (50-200 ms) respectively [7,8]. These consistencies validate the setup's accuracy. For improvement, we suggest fully integrating an SDR transmission stick into GNU Radio, allowing cost-effective hardware use while relying on a single, user-friendly software platform. This optimization could reduce development costs and expand potential applications. This project demonstrates that a functional MR signal reception path can be built affordably by combining cost-effective hardware with open source, user-friendly software which was confirmed by initial proof-of-principle T2 measurements. The required software, GNU Radio, is freely accessible and easy to operate, making this setup accessible even for users with limited technical expertise. This approach opens new possibilities for low-cost NMR applications, making the technology more accessible even beyond the MR community.
Lilli BREUER (Lüdenscheid, Germany), Maurice RÜGER, Tobias KRAATZ, Helena NAWRATH, Amir MOUSSAVI, Jens GRÖBNER
11:00 - 12:30
#47785 - PG323 Impact of decreased antenna shield spacing on antenna design and efficiency.
PG323 Impact of decreased antenna shield spacing on antenna design and efficiency.
Multichannel transmit systems in MRI at 7 T improve excitation homogeneity [1,2], preferring dipoles [3] and meander elements (ME) [4] as transmit elements. Implemented as local coils, they require considerable scanner space. To preserve patients and equipment space, an integrated 32-channel array [5] was installed at the DKFZ. Newer Siemens 7T MRI systems, however, employ smaller gradient coils, reducing installation space by two-thirds. This constrain affects antenna performance, as the H-field magnitude depends on RF shield height [6].
This study evaluates the impact of limited space on transmit capabilities, comparing the performance of ME of the current 32-channel system at different heights over the RF shield with a modified ME and bent dipole (BD) [7].
Modelling and EM simulations were done using CST Studio Suite (CST AG, Darmstadt, Germany). The transmit elements were designed for a Siemens Magnetom 7T system (Siemens Healthineers, Erlangen, Germany) equipped with a SC72 gradient coil at 12 mm distance to the bore liner.
The original ME was simulated at 18 mm distance to the RF shield (h₁) and at 9 mm (h₂), to meet spatial constrictions, along a modified ME and BD designs (Fig. 1). Central feeding was co-simulated using a λ/2 balun (330 mm length, 50 Ω impedance) and a matching network with lumped capacitors. All resonant elements and RF shields were simulated as PECs. A lossy 1-mm thick RO4350B substrate was used. The elements were placed below a phantom (330x310x90 mm³) of tissue-simulating liquid (ε'r: 46, σ: 0.87 S/m) at distances of 20 mm (d₁) and 150 mm (d₂). Power (B₁⁺/√Pacc) and SAR efficiency (B₁⁺/√SAR) inside the phantom and coupling were evaluated, with results normalized to 1 W (RMS) of accepted power. Table 1 shows antenna dimensions and S₂₁ coupling under varied loading. BDs required a small capacitive area for a smaller Lc to maintain central field distribution. At d₁, coupling remained near −20 dB for all elements, while it decreased at d₂ with reduced RF shield spacing. The original ME placed at h₁ showed the highest coupling (−4 dB), reduced to −9.89 dB at h₂. The modified ME had the lowest coupling, reducing S₂₁ by 50% compared to unmodified ME. All BDs maintained <−10 dB values.
Contrary to prior work [6], shield proximity improved overall efficiency at short phantom distances (Fig. 2). BD showed higher B₁⁺/√Pacc and B₁⁺/√SAR peak values than the MEs, despite reduced z-axis coverage. At d₂, the element farthest from the RF shield showed superior power efficiency, opposite to the trend at d₁. In general, MEs designs outperformed BD, showing 33% lower B₁⁺/√Pacc.
Antennas near the RF shield performed better in superficial phantom layers at d₁ (Fig. 3). Beyond 60 mm depth, all designs performed similarly. At d₂, MEs exhibited superior power efficiency, with no significant SAR efficiency variations. Reflections at the phantom edge caused minor efficiency curve fluctuations due to phantom size. The impact of RF shield spacing on transmit performance was evaluated for ME and BD at 7 T. In MEs, longer meanders increase electrical length, requiring lower end capacitance (Ce) for compensation [4]. In the ME with short meander, reduced Ce was also required to maintain central field distribution, suggesting a combined effect of increased electrical length from closer RF shield proximity and a thinner microstripline. At larger phantom distances, MEs located farther from the RF shield showed higher power and SAR efficiency. As demonstrated in earlier work [6], increased shield spacing improves field penetration. However, the opposite trend was observed with a close load, suggesting that load proximity also influences field behavior.
RF shield proximity improved transmission performance in superficial layers of the phantom, while efficiencies beyond 60 mm depth remained similar. At d₂, ME greatly outperform BDs. SAR efficiency remained similar within the phantom, proving to be independent of shield distance.
Coupling was reduced for d₁ due to EM field concentration between coils and phantom, limiting field interactions with neighboring elements. At longer load distances, reduced RF shield spacing significantly reduced coupling across all elements. Lower coupling in all BDs indicated less power transfer to adjacent elements, due to lower B1+ efficiency. Spatial limitations in MRI systems requires evaluation of transmit elements placed closer to the RF shield for next generation transmit arrays. ME demonstrated strong performance under all simulated conditions. At short phantom distances, MEs near the RF shield demonstrated both improved power and SAR efficiency, while larger shield spacing is preferred when transmitting at greater load distances. Intrinsic decoupling was overall superior for thinner ME. No gain was observed by using distributed capacitors for BDs.
Ultimately, ME proved to be a strong candidate for implementation into arrays for future MRI multichannel transmit systems.
Andrea PINO RAMOS (Heidelberg, Germany), Mark E. LADD, Stephan ORZADA
11:00 - 12:30
#47799 - PG324 Investigation of mechanical stability of bore liner for a high-count UHF RF transmit array.
PG324 Investigation of mechanical stability of bore liner for a high-count UHF RF transmit array.
At 7 T, multichannel transmit arrays improve B₁⁺ homogeneity [1,2] typically using local transmit coils, which require substantial bore space within the scanner bore liner. To preserve space for patients and equipment, an integrated 32-channel array was installed at the DKFZ [3]. Newer Siemens 7T MRI scanners employ smaller gradient coils, reducing installation space by two-thirds. Reduced spacing between transmit elements and RF shield alters H-field distribution [4], affecting transmission performance.
To enable array integration under these constraints, structural modifications to the bore liner constructed from fiber-reinforced plastic (FRP) were explored. Mechanical analysis focused on stress distributions, using Maximum Principal Stress (MPS) and von Mises Stress (SvM) [5], criteria suited to evaluate structural response in FRPs [6].
Alternative bore liner designs with integrated antenna slots were analyzed using finite element analysis (FEA) under conservative loading.
The modelling and finite element analysis (FEA) structural simulations were done with ANSYS Mechanical (ANSYS, Inc., Canonsburg, PA, USA) using a bore liner model from Siemens Magnetom 7 T system (Siemens Healthineers, Erlangen, Germany). Material data was provided by Maschinenfabrik Reinhausen GmbH (Reinhausen, Regensburg, Germany). The bore liner consists of E-glass/epoxy (roving, 79.5 wt.-% glass), with mechanical limits set by IEC 61462: 60 MPa axial and 120 MPa tangential stress. E-Glass Wet was simulated for its similar anisotropic properties.
To accommodate the 32-channel array (Fig. 1), 32 antenna-sized slots (250×50 mm²) were introduced with spacing of 28 mm (xy-plane) and 50 mm (z-axis). Load simulations accounted for a combined mass (patient, table, and dedicated receive array) of 2000 N, with 1000 N applied per rail for symmetric loading. Four fixed supports (110×96 mm²) constrained the structure. An adaptive mesh was refined near critical regions.
Three designs were analyzed: original, slotted with sharp edges, and slotted with 24 mm-radius edge rounding. For all models, MPS and SvM assessed the static structural response. Fig. 2 and 3 show simulation stress distribution in the bore liner, viewed in lateral and base sections. Table 1 shows the maximum FEA values obtained for each case.
For stress analysis, a MPS of 2.89 MPa was obtained for the original bore liner (A). Introducing 32 slots with sharp-edged corners increased the stress nearly twice compared to the original design, increasing to 7.18 MPa. Applying a 24 mm radius to the 32 slots (C) reduced the value to 5.84 MPa.
In SvM calculations, the original structure (A) exhibited a stress value of 2.83 MPa. Introducing 32 slots with sharp corners increased the stress to 6.8 MPa, nearly 2.5 times higher than the original. Applying a 24 mm radius at the slot base reduced the stress to 5.81 MPa, representing a decrease of approximately 1 MPa compared to the sharp cornered design.
Across all models, the stress values obtained in both analyses were well below the limits under internal pressure of 60 MPa and 120 MPa. The highest stress concentrations were consistently located near the borders of the supporting rails, with pronounced intensity in the central base area of the structure (Fig. 3 A-F). The original bore liner consistently exhibited the lowest values across all failure metrics, serving as a comparison baseline. Introducing sharp-edges slots significantly increased stress, nearly tripling peak failure values due to geometric discontinuities acting as stress concentrators. These results align with previous work [5,7], showing that in FRPs stress concentrates around cut-out areas. Rounding the slots edges notably reduced peak stress, indicating improved load distribution, resistance to failure and reducing the stress concentration factor. Replacing sharp with rounded edges redistributes stress more evenly, consistent with established strength optimization strategies [6].
All stress levels remained below IEC 61462 limits, with a maximum of 7.18 MPa (~10% of max. stress limit), well within the E-glass allowable range. This suggests that the design modifications are likely to maintain structural integrity under the applied load conditions. Top region of the liner showed minimal stress (in contrast to stress concentration along rail borders), suggesting it as a viable area for RF coil integration under reduced mechanical loading. High-count integrated arrays are incompatible with the spatial constraints of new Siemens 7 T systems. Structural modifications to the bore liner were evaluated to enable array integration and preserve performance while maintaining mechanical stability. FEA simulations showed that refining the slot geometry with rounded transitions effectively decreased principal stresses concentrations and improved load distribution.
These findings support the feasibility of slot integration, provided that key geometric features are optimized for mechanical stability.
Andrea PINO RAMOS (Heidelberg, Germany), Mark E. LADD, Stephan ORZADA
11:00 - 12:30
#47743 - PG325 Loop Inductive Coupling for Efficient Solenoid Matching in Low-Field MRI study.
PG325 Loop Inductive Coupling for Efficient Solenoid Matching in Low-Field MRI study.
Impedance matching plays a critical role in optimizing the performance of radiofrequency (RF) coils in Magnetic Resonance Imaging (MRI), especially under low magnetic field conditions where the signal-to-noise ratio (SNR) is inherently limited[1, 2]. Traditional matching circuits rely on variable capacitors, which are not only sensitive to loading conditions but also contribute to noise and increase setup complexity. In this project, we explore an alternative, passive method of impedance adaptation using loop inductive coupling to simplify the matching procedure of a solenoid coil. The objectives are component cost and complexity reduction, and SNR improvement, in the context of 0.1 T MRI exploration.
Experiments were conducted on an open 0.1 T vertical-field electromagnet (EAR 50 L, Drusch, France) (Fig. 1). This resistive magnet is powered by a high-stability power supply and water-cooled during operation. This system also embeds magnetic field gradient coils and shimming coils (B0 homogeneity correction on 3 first order and 5 second order components).
The transmit-receive solenoid coil[3] consists of a 12 turns solenoid (125 mm x 65mm) made with 6 mm cross-section copper tube. It is tuned to the Larmor frequency thanks to three parallel 22 pF ceramic capacitors and two 0.8 – 8 pF variable capacitors directly soldered onto it. To facilitate precise tuning prior to acquisition, resonance is achieved by adjusting the B0 field strength through modulation of the magnet power supply current.
A secondary non-resonant inductive loop, mechanically repositionable along the solenoid long axis, is used to adjust impedance matching instead of an additional circuit as traditionally done[4]. Matching efficiency was evaluated using a Vector Network Analyzer (ZNB4, Rohde & Schwarz). Two loop positions were compared: Pos0 = the loop is positioned at the solenoid entrance, and Pos1 = the loop is positioned around the solenoid at a location minimizing antenna’s return loss - S11 (dB). S11 and quality factors were measured with and without the phantom load (a cylinder filled with tap water mixed with copper sulfate (CuSO₄) to shorten relaxation times).
MRI data were acquired on the phantom using three different sequences: 2D Gradient Echo (TE = 3.8 ms, TR = 202 ms, FA = 60°, FOV = 250mm2, matrix = 64 x 64), 2D Fast Spin Echo (TE = 5.06 ms, TR = 500 ms, ETL = 5, FOV = 250mm2, matrix = 64 x 64), and 3D Spin Echo (TE = 5.78 ms, TR = 500 ms, FOV = 250mm2, matrix = 64 x 64). NMR signal profiles, noise background and nutation curves were obtained with standard one pulse sequence and were processed with MATLAB. The mean signal and the noise standard deviation were extracted from regions of interest defined using the graphical tool integrated into the system console (Chameleon 4, RS2D, France). The SNR was then calculated by accounting for the Rayleigh distribution of noise in magnitude images[5] as follows:
SNR= Mean(Signal)/(SD(Noise))× √(2- π/2) (1) At Pos1, the loaded coil achieved a minimum S11 of –40.68 dB at 4.54 MHz vs -8dB at Pos0 (Fig. 2). Q factor calculated as the ratio between Qunloaded and Qloaded was 1 in both cases. NMR signal peak showed good field homogeneity in both cases (<13 ppm). Signal intensity of one pulse acquisition increased by 59.4% in Pos1 compared to Pos0 (Fig. 3.A). Standard deviation of noise acquisition also increased by 27.6% though. Nutation experiments confirmed same excitation pulse calibration led to better signal reception in Pos1 (Fig. 3.B). Image acquisitions showed significant enhancement in signal intensity at Pos1, with decreases or only minor increases in background noise depending on the evaluated slice, resulting in an overall improvement of SNR across all slices for the three imaging sequences (Table 1). The use of an inductively coupled loop for impedance matching presents multiple advantages for low-field MRI systems. It eliminates the need for noise-prone electronic components, enables rapid and user-friendly matching adjustments, and reduces development costs. Additionally, this method allows flexible adaptation of coil configurations to varying subject morphologies or phantom geometries that lead to different filling factors, improving overall signal capture. The measured SNR gains demonstrate the method’s clinical potential aligning with current trends in developing lighter and cost-effective low-field MRI systems. We demonstrate the feasibility of achieving highly efficient and flexible impedance matching for a solenoid coil using passive inductive coupling in a 0.1 T MRI system. This approach significantly enhances SNR without electronic components, reduces system complexity, and opens new perspectives for the development of accessible point-of-care MRI technologies.
Marie-Anaïs PETIT (Geneva, Switzerland), Redha ABDEDDAÏM, Marc DUBOIS, Delphine BECHEVET
11:00 - 12:30
#47625 - PG326 Design and Simulation-Based Optimisation of a Combined ¹H/²³Na/³⁹K RF Coil for 7T MRI.
PG326 Design and Simulation-Based Optimisation of a Combined ¹H/²³Na/³⁹K RF Coil for 7T MRI.
Combining proton(¹H), sodium(²³Na), and potassium(³⁹K) imaging offers significant clinical potential for investigating Na/K pump deficiencies associated with conditions like epilepsy, stroke, or cancer(1–3). While triple-frequency coils(¹H/²³Na/³⁹K) have been demonstrated for preclinical application(4), no equivalent for human has been reported to date. Previously, we presented separate coil designs: a 16-channel ²³Na-loop/¹H-dipole array(5), and a ³⁹K birdcage coil(6) for 7T MRI. In this study, we introduced a novel triple-resonant RF coil for human head imaging, integrating our previously developed coils. EM simulations were performed to optimise coil structure in terms of S-matrices and B₁⁺ performances and assess RF safety with SAR₁₀g using a human head model.
The triple-frequency coil combines previously developed 16ch ²³Na-loop/¹H-dipole array(Fig.1a) and ³⁹K birdcage coil(Fig.1b). The ³⁹K birdcage was inserted into the existing ²³Na/¹H structure, forming a double-layered coil. Four orientations were simulated to identify the optimal configuration(Fig.1c): 1.0° rotation-birdcage sources aligned symmetrically with dipoles;2.22.5° rotation-birdcage sources placed midway between dipoles;3.45° rotation-birdcage sources realigned with dipoles;4.dipoles directly mounted onto the birdcage instead of the loop layer.
Finite-difference time-domain(FDTD) simulations(Sim4Life 8.2,ZMT,Switzerland) were performed with a spherical phantom(d=15 cm,εᵣ=81,σ=0.95 S/m). Dipoles were modelled as centre-shortened structures on FR-4 substrates(εᵣ=4) with lossy metal(σ=5.8×10⁷S/m). Loop coils and birdcage elements were modelled as perfect electric conductors (PEC) with zero thickness surfaces. Alternate loop coils were elevated by 8 mm to facilitate voxelisation. An RF screen(PEC) was also included. Individual nuclei simulations (¹H:297.2 MHz, ²³Na:78.6 MHz, ³⁹K:13.9 MHz) were performed separately, with other coil elements present but inactive. Co-simulation (OptenniLab 5.2,Finland) adjusted lumped element values for tuning and matching. S-matrices and circularly-polarised(CP) B₁⁺ maps were extracted to assess coil performance (Figs.2,3). Separate simulations were performed for single-nuclei elements for comparison.
Following phantom optimisation, RF safety was evaluated using the Duke head model at 2-mm iso resolution(Fig.4a). B₁⁺ and SAR₁₀g maps were interpolated to 1-mm iso resolution, normalised to 1W total input power, and exported to Matlab for analysis. S-parameters for all setups were compared to independent single-nuclei coils (Fig.2). Setup 4 (dipoles on birdcage) provided optimal 1H performance, achieving the lowest reflection (Sᵢᵢ=-25.6 dB) and best decoupling (Sᵢⱼ<-13.4 dB). ²³Na and ³⁹K coil performances were consistent across setups, closely matching individual coil benchmarks.
B₁⁺ maps indicated significant orientation dependence for ¹H imaging(Fig.3). Setups 1–3 demonstrated reduced B₁⁺ efficiency(17–54% decrease in mean B₁⁺) compared to independent ¹H-dipole array, while Setup 4 markedly improved mean B₁⁺ by 66.7%. For ²³Na, all setups showed increased B₁⁺ of 43–68% with minimal orientation dependence. The ³⁹K coil showed consistent increase of 17–20% in B₁⁺ across all setups.
Due to superior performance across all nuclei, Setup 4 was selected for safety evaluation. Duke head simulations confirmed good matching(Sᵢᵢ<-15 dB) and isolation(Sᵢⱼ<-7 dB). B₁⁺ on the central slice and maximum intensity projection(MIP) for SAR₁₀g are shown in Fig 4. Maximum SAR₁₀g values were reported as 0.25W/kg(¹H), 0.12W/kg(²³Na), and 0.24W/kg(³⁹K). In this study, we proposed a compact triple-resonant coil integrating ¹H, ²³Na, and ³⁹K imaging elements at 7T. Setup 4 provided the optimal balance of coil performance, demonstrating excellent S-parameters and efficient B₁⁺ fields. This advantage was most pronounced for ¹H, likely due to dipoles mounted directly on the birdcage, reducing shielding effects in other setups. Notably, setups 1 and 3 yielded similar B₁⁺ magnitudes but differed in spatial orientation, with the ¹H field pattern clearly rotating by 45° between these configurations.
In Duke simulations, the coil showed maximum SAR₁₀g approximately 30% higher for ¹H compared to previously reported values for separate ¹H-dipole array, concentrated mainly near the ears due to their proximity to dipole elements. SAR₁₀g levels for ²³Na and ³⁹K remained comparable to previously validated coil designs, indicating minimal additional risk introduced by combining elements. However, additional safety validation remain necessary with human evaluations. We have demonstrated a promising initial design and simulation-based optimisation of triple-resonant(¹H/²³Na/³⁹K) RF coil at 7T. The optimised coil(Setup 4) achieved excellent multinuclear performance with SAR evaluated in simulations using a realistic human head model, highlighting its potential clinical utility. Future work will focus on coil fabrication, bench testing, and safety validation.
Menglu WU (London, United Kingdom), David W. CARMICHAEL, Özlem IPEK
11:00 - 12:30
#47836 - PG327 Optimizing Signal-to-Noise Ratio at 7T MRI Using Algorithmic Selection in a 16-Channel Receive Array.
PG327 Optimizing Signal-to-Noise Ratio at 7T MRI Using Algorithmic Selection in a 16-Channel Receive Array.
High-field MRI systems benefit from parallel receive coil arrays, which can significantly enhance the signal-to-noise ratio (SNR). Current state of the art research increase the number of coils as a means to increase the SNR [1-4], where the coil geometry is not optimized. One of the primary challenges in multichannel receive arrays is interelement decoupling. This decoupling is a key limiting factor in optimizing both the individual coil geometry and the overall array configuration for maximum signal-to-noise ratio (SNR). Traditional decoupling techniques, such as partial coil overlap and preamplifier decoupling, inherently constrain the flexibility of coil design.
Recently, novel coil designs based on transmission line modifications [5, 6] have been introduced as self-decoupled elements. These coils maintain effective decoupling and performance even when their shape deviates from the conventional circular form, which makes them a good candidate for optimized coil geometry.
This study proposes a novel approach to designing receiving coil array geometries using an algorithm to find the optimal coil shape combination that maximizes SNR in user defined regions using a limited number of coils.
420 coils were modeled using 100 line-segments per coil. The coils were placed on a cylindrical plane 2 cm from the phantom. All coils have a wire length of 31.415 cm and consist of various shapes (squares, triangles, vertical and horizontal rectangles, circles and figure eights). A homogeneous cylindrical phantom was modeled (height = 20 cm, radius = 10 cm, σ = 0.55 S/m, ε = 80, ⍴ = 997 kg/m^3) with a voxel resolution of 6mm. The electric field (E) and magnetic flux density (B) were simulated per coil individually in the phantom using the MARIE solver [7] in MATLAB.
The loss matrix is calculated as Q = dV ∑ σ(r)E(r)E(r)^H, where dV is the voxel volume. The B1 fields are calculated as B1- = (Bx - 1i*By)/√2 and B1+ = (Bx + 1i*By)/√2. The SNR per voxel is then calculated as SNR(r) = B1- x/√(x^H*Q*x), where x is a weight vector between the coils which is to be found for maximum SNR. The solution to this optimization problem is well known and will not be discussed here.
2 regions are defined in the phantom, the periphery (|z| < 5 cm, r > 8 cm) and the center (|z| < 5 cm, r < 6 cm), as shown in Fig. 2 (a).
The optimization goes as follows. First a region of interest is chosen (periphery or center), then the average SNR of all coils is in this region is calculated individually. The one with the highest mean SNR is added to the array. Next all non selected coils are individually tried in combination with the existing array. The one resulting in the highest mean SNR is added to the array. This process is repeated until the array consists of 16 coils (Fig. 1).
As a comparison a conventional equispaced 16 coil circular loop array is also examined. Loops were placed in two rows (Fig. 2(b)) and each loop was of 10cm diameter. The found geometries of the optimized arrays are shown in Fig. 2 (c-d).
Calculations of the SNR, portrayed in Fig. 3, demonstrate a significant improvement for the optimized coil arrays compared to circular loop array. The periphery optimized array shows a mean increase of 67% of the mean periphery SNR and a 15% decrease in the mean center SNR compared to the loop array. The center optimized array shows a mean increase of 17% of the mean periphery SNR and a 6% increase of the mean center SNR compared to the loop array. With the approach shown in this manuscript, we demonstrated that improvement in peripheral SNR can be achieved by optimizing the coil geometry and its position within array.
While algorithmic optimization outperforms conventional loops, it does not guarantee a globally optimal solution, an improved algorithm might yield better results. Future research should include algorithm optimization strategies.
The assumption of complete decoupling does not hold for all coil types, in this research transmission line coils are assumed. If other coil types are used additional decoupling hardware might be necessary for the SNR to be improved using these coil geometries.
Further improvement in SNR can be achieved by considering specific clinical applications, such as optimizing for a specific part of the brain. Future research will focus on realistic human body models. Also, as a future work optimization would be performed on higher channel number such as 32, 64 or even higher. Algorithmic optimization has been shown to improve SNR at 7T using a 16-channel receive coil array. This approach provides a promising avenue for enhancing image quality in high-field MRI systems.
Yannick LARET (Eindhoven, The Netherlands), Bart ERICH, Irena ZIVKOVIC
11:00 - 12:30
#47661 - PG328 RF array coil and passive dipoles for spine MRI at 7T.
PG328 RF array coil and passive dipoles for spine MRI at 7T.
MRI is a modality of choice for spine imaging as it allows optimal visualization of soft tissues. In the last decades, Ultra-High-Field scanners operating at 7 T have been developed with the aim of improving signal-to-noise ratio (SNR) and resolution. However, these technical advantages came at the cost of new challenges such as increase of the specific absorption rate (SAR) and inhomogeneities of the radiofrequency field B1 [1]. These inhomogeneities are mainly related to the shorter operating wavelength and have changed the concept of optimized RF coils. Indeed, at UHF, volumetric coils might not be optimal and surface coils adapted to a given region of interest (ROI) might be preferred. Our target area, the thoracic or lumbar spine, is 25-30 cm long, less than 1 cm in diameter, and lies at 4-6 cm under the skin [2]. The optimal coil design for this region for UHF MRI is not yet known [3].
We propose a coil array specifically adapted for the thoracic and lumbar spine geometry. It is made of 8 loops that can be independently controlled in amplitude and phase, thus allowing for B1 shimming and parallel transmit [4][5]. While some studies have shown the interest of using active dipoles to improve in depth transmission [6], we provide evidence that B1 intensity and homogeneity in the ROI can be improved by adding carefully placed passive dipoles to our design. In addition, maximum SAR can be reduced thereby ensuring a better transmit efficiency.
Eight 10 cm diameter loops, segmented with three 2.7 pF capacitors were positioned in two rows and overlapped to minimize inter-element coupling. Each loop was tuned and matched to the Larmor frequency of 297.2 MHz and connected to the MRI scanner through an interface box (Stark Contrast, Germany). In addition, two metallic stripes of 600 mm length, 12.5 mm width and segmented in their middle with 10 pF capacitors were placed under the outer edges of the coil array in order to act as passive dipoles.
Simulations of this setup were run on the time-domain solver of CST Studio Suite with a Finite Integration Technique.
In both numerical simulations and experiments, the array was placed on a foam spacer 10 mm above the 2 dipoles which were located 10 mm above a 480x350x175 mm3 phantom filled with a dedicated body liquid (εr=58,2 and σ=0,92 S.m-1). (Fig. 1)
Experiments were conducted at 7T (Magnetom TERRA, Siemens Healthineers, Erlangen, Germany). B1+ maps were acquired with a turboFLASH sequence [7]. A reference static excitation was selected, with each channel set to the same amplitude and a 180° phase shift between the two rows in order to produce constructive currents in the center of the array [8].
The performance of the array and the two dipoles combined was compared to the performance of the array alone. To do so, a region of interest corresponding to the hypothetic position of the spinal cord was defined as a 300x3x3 mm3 box lying 5 cm deep into the phantom. Simulations indicated that the addition of the 2 dipoles improved the average amplitude of the field by 21% and reduced the relative standard deviation by 34%. The maximum SAR (in the whole phantom) was reduced by 26% and thus the transmit efficiency (defined as the ratio between average amplitude and square root of maximum SAR) was improved by 40% in the ROI. (Table 1)
Experiments confirmed that the 2 dipoles were effectively focusing the field in the central region (Fig. 2). The average B1+ field was improved by 25% while the Relative Standard Deviation was reduced by 15% in the ROI (Table 1). In the case of the array alone, the current patterns applied in the loops to generate a bright B1+ field in the ROI also lead to undesirable illuminations under the outer edges of the array. By adding the two dipole-like elements and tuning them so that their surface currents become out of phase with those of the outer edges of the array, we can offset the lateral contributions and thus cancel the B1+ field outside of the desired area (Fig. 3)
The experimental results will need to be reproduced and tested for different ROIs to determine error margins and identify possible causes of mismatch with the simulations. Our results illustrate that through a better distribution of the transmit field, the addition of passive dipoles can improve the classic coil array design. Simulations indicate a gain in both B1+ homogeneity and intensity, and also predict a reduction of the maximum SAR. Experiments further confirm this working principle and show that the dipoles effectively focus the field on the ROI. The in vivo assessment of this prototype is under progress.
Hugo AMAT (Marseille), Aurélien DESTRUEL, Amira TRABELSI, David BENDAHAN, Virginie CALLOT, Stefan ENOCH, Redha ABDEDDAIM, Marc DUBOIS
11:00 - 12:30
#47887 - PG329 A Multi-Vendor compatibility Study of a Wireless RF Coil for Breast Imaging.
PG329 A Multi-Vendor compatibility Study of a Wireless RF Coil for Breast Imaging.
Development of radiofrequency (RF) coils for clinical MRI focuses on enhancing image quality, patient safety, and comfort. Wireless RF coils operating by inductive coupling with the body birdcage coil (BC) are an alternative to conventional cable-connected RF coils [1, 2, 3]. These coils offer significant advantages over traditional cable-connected coils, including reduced electromagnetic interference and improved patient comfort since no thick cables are used. However, since wireless coils are usually operating in transceiver (TxRx) mode, it is necessary to perform manual calibration of the reference Tx voltage to avoid inhomogeneity of the B1+ field [2]. It complicates clinical workflow and increase scan time.
To address these limitations, we have developed a concept of a wireless receive-only RF coil (Rx-only) for 1.5T MRI [1], specifically designed for breast imaging where dedicated multi-channel coils are often unavailable across different vendor platforms. The design is based on a Helmholtz coil with two passive diode traps for detuning. This means that imaging setup with wireless coil is performed in Tx mode similarly to traditional setup with cable connected Rx coils. In receive mode, the Rx-only wireless coil is tuned and is able to detect the MR signal, which then is injected to Rx-chain of MR scanner via inductive coupling with BC. Therefore, it makes possible to use this Rx-only coil without specific adjustments [1, 2].
In this study, we evaluate the multi-vendor compatibility of the proposed Rx-only wireless coil through experimental validation with 1.5T (Larmor frequency 63.8 MHz) MRI systems of major vendors (GE, Siemens, Philips). Results demonstrated the coils ability to perform MR imaging without disrupting standard imaging protocols, offering a practical solution for clinical MRI across different vendors.
The experimental study was performed using our previously developed Rx-only coil prototype [1]. This is a Helmholtz coil incorporating two passive LC traps (with inductance of 67 nH and a capacitance of 89.5 pF) inserted into the gap of the coil conductor (Fig.1d). Each inductance is connected in series with a pair of back-to-back Shottky diodes. During the transmit mode, forward-biased diodes activate the traps, shifting the resonance frequency away from the Larmor frequency. In the receive mode, reverse-biased diodes disable the traps, that allows to operate Rx wireless coil in receive mode.
Experimental study was performed on three different 1.5T MRI systems using the same spherical phantoms filled with a NiCl2 water solution. Siemens Espree Magnetom was used to acquire phantom images and flip angle (FA) distributions using the double-angle method [4] (TR/TE = 9000/4.76 ms, matrix = 128×128 mm²). For Siemens system SNR calculations were performed with FA = 90°. FA mapping employed gradient-echo sequences with FA of 40° and 80° for the double-angle reconstruction. GE Optima 360 system was used for comparative analysis of the Rx-only coil versus 8-channel HD Breast coil using identical scan parameters. On the Philips Achieva system, qualitative image assessment was performed against a SENSE breast 7-channel coil. All experimental setups for each vendor platform are shown in Fig. 1 a-c. Figure 2a shows the phantom image acquired with the Rx-only coil on the Siemens system, demonstrating an SNR of 650. In comparison, the BC-only configuration achieved a lower SNR of 115. For the GE scanner, the phantom image acquired with the Rx-only coil (Fig. 2b) was compared with 8-channel HD Breast coil (Fig. 2c). Additionally, Philips system evaluations of the wireless Rx–only coil (d) and 7-channel SENSE coil (e) are presented. On GE and Philips systems it is not possible to disable the transmit coil by software, so it is not possible to obtain correct noise maps for SNR assessment. Figure 3 presents the FA distribution maps obtained from Siemens scanner for: (a) the Rx-only coil and (b) BC-only configuration. The results demonstrate successful multi-vendor compatibility of the Rx-only coil across three MRI vendors. The design overcomes vendor-specific limitations like absence of manual voltage calibration required by Tx/Rx wireless coils, particularly critical for GE and Philips systems. The wireless coil provides consistent performance without protocol modifications and has much lower cost. The wireless receive only coil provides multi-vendor compatibility with 1.5T MRI-systems eliminating manual calibration of Tx voltage. This design offers a cheaper and simpler alternative to conventional Rx coils.
Aleksandr FEDOTOV (Saint-Petersburg, Russia), Pavel TIKHONOV, Georgiy SOLOMAKHA, Alexandr KOZACHENKO, Anna HURSHKAINEN
11:00 - 12:30
#45925 - PG330 Traveling-Wave MRI Extends Surface Coil Coverage.
PG330 Traveling-Wave MRI Extends Surface Coil Coverage.
Conventional surface coils in MRI are inherently limited to a field-of-view (FOV) constrained by their physical dimensions. The traveling-wave MRI (twMRI) approach overcomes this limitation by enabling large-FOV imaging, as previously demonstrated [1]. In this work, we present an enhanced twMRI system combining: (i) a parallel-plate waveguide (PPWG) that eliminates cutoff frequency restrictions, and (ii) a bioinspired surface coil design [2]. Building on prior work showing metamaterials can improve signal-to-noise ratio (SNR) in this configuration [3], we address a fundamental limitation of traditional twMRI implementations. The primary objective of this study was to experimentally validate that our twMRI-PPWG system with a bio-inspired surface coil can achieve an extended FOV beyond the coil's physical dimensions while maintaining high image quality.
A bio-inspired surface coil, as described in [2], was employed for RF signal transmission. The metamaterial structure, designed in a nonagonal configuration, consisted of nine copper strips (500 mm × 10 mm × 35 μm, σ = 5.96 × 10⁷ S/m) laminated onto FR4 substrates (ϵ = 4.35, tanδ = 0.008, 1 mm thickness). Each strip featured a periodic array of 49 C-shaped unit cells (5 mm diameter, 3 mm gap width, 2 mm conductor width; Fig. 1.b) arranged linearly in a 1×49 configuration (Fig. 1.a). Phantom imaging experiments were conducted using a saline-filled parallel-plate waveguide (PPWG) with the metamaterial positioned inside (Fig. 1.c). The bio-inspired surface coil was placed externally, aligned parallel to the waveguide plates to maximize coupling. To validate the approach, sagittal phantom images were acquired with a standard gradient-echo sequence (TE/TR = 4.39/200 ms, FOV = 60 × 60 mm², matrix = 256 × 256, flip angle = 45°, slice thickness = 1 mm, NEX = 4). For comparison, additional images were obtained using both the bio-inspired coil and an in-house birdcage coil (length/diameter = 5 cm/4 cm, 4 rungs). All experiments were performed on a 7T/30 cm Bruker scanner (Bruker BioSpin MRI GmbH, Germany) and cylindrical phantom (diameter/length = 4 cm/ 10 cm) filled with saline solution. Phantom images acquired with the nonagonal metamaterial confirmed the feasibility of the approach (Fig. 2b). Signal-to-noise ratio (SNR) analysis yielded the following values: SNR(nano) = 99, SNR(inhBC) = 106, and SNR(bio) = 115. A profile comparison of the twMRI and the in-house BC coil data (Fig. 2.a) revealed similar performance between the two methods. However, the twMRI signal exhibited a gradual decline with distance from the coil, reaching a maximum decrease of 27% relative to the initial signal. Notably, the twMRI image avoided the pronounced signal drop observed at the end rings of the in-house BC coil profile (Fig. 2.a), demonstrating a key advantage. Furthermore, the phantom images (Fig. 2.b–c) indicated that the twMRI provided greater coverage than the bio-inspired surface coil. This is a rather encouraging result, as the signal was transmitted 58 cm away from the phantom's location. Our results demonstrate that the twMRI approach achieves a significantly larger FOV than conventional surface coils, exceeding the coil’s physical dimensions while maintaining competitive image quality. This work represents a key advancement in the practical implementation of twMRI, enabling expanded anatomical coverage without compromising sensitivity—a critical requirement for many imaging applications.
Sergio SOLIS-NAJERA, Jelena LAZOVIC, Saul RIVERA DE LA LUZ, Alfredo RODRIGUEZ (Mexico City, Mexico)
11:00 - 12:30
#47600 - PG331 Metasolenoid resonator for controlling magnetic field in 3T MRI.
PG331 Metasolenoid resonator for controlling magnetic field in 3T MRI.
Over the past decade, there has been an increasing interest for radiofrequency devices dedicated to imaging of specific anatomical areas, such as breast, wrist etc... Metamaterial resonators at 1.5T [1,2] and ceramic resonators at 3T [3] have already been shown to be effective for targeted MRI applications but experimental data related to a metamaterial volumetric resonator operating at 3T are missing. Here we propose a passive metamaterial resonator designed for a 3T MRI scanner. In addition, we analyzed the influence of two birdcage excitation modes.
The design of the metasolenoid illustrated in Figure 1 was inspired by a previously reported volumetric resonator operating at 1.5T [4]. It consists in four PCB plates soldered together in a rectangular box shape. Each plate contains 10 etched copper strips so that the entire device is composed of 10 split-loop resonators, with a gap on each plate for capacitors. Its resonance frequency was tuned to 123.2 MHz by adding three 3.3 pF and one 2.7 pF ceramic capacitors (Johanson Technology and Knowles) in the gaps of each SLR. The resonance frequency was also characterized by connecting a non-resonant loop antenna connected to a vector network analyzer (MS2026C, Anritsu). A tissue-mimicking liquid (MVG, France) with a relative permittivity of 61.9 and a conductivity of 0.8 S/m was used to create the phantom by filling a 2.4 L glass bottle. The experiments were performed in a 3T Vida MRI scanner (Siemens Healthineers, Erlangen, Germany) using a whole-body birdcage coil (BC) for both signal transmission and reception. Flip angle (FA) maps were acquired in the sagittal orientation using a pre-pulse RF with a TFL readout sequence [5], TR/TE = 7030/1.8 ms, FA = 8°, Sinc pulse = 80°, FOV = 289 x 289 mm, matrix size = 128 x 128 and 13 slices of 10 mm thickness. The reference voltage was calibrated for each set of measurements. The birdcage excitation was used as a reference. We performed measurements with two different polarizations : circular polarization (CP) with both birdcage ports excited in quadrature and linear polarization (LP) by selecting only the birdcage port that couples to the metasolenoid resonator. The reflection coefficient shown in Figure 2 was measured using a non-resonant measurement loop in order to estimate the resonant frequency of the metasolenoid. A sharp dip could be observed near the Larmor frequency when the metasolenoid was not loaded, indicating that the metasolenoid was properly tuned. Accordingly, for the loaded metasolenoid, a reduction of the resonance quality factor was observed. The experimental setup and FA maps are shown in Figure 3. Panels B–E show the FA map in the mid-slice of the phantom. The FA averaged in the ROI and the reference voltage are given for each measurement. Panels B–C show the Birdcage reference coil for CP and LP excitations, respectively, panels D–E show the results with the meta-solenoid. Table 1 presents a summary of all results. As compared to the reference birdcage-only experiment, the addition of the metasolenoid was able to significantly reduce the reference voltage required to achieve the target FA in the phantom ROI. Transmission efficiency was calculated as the ratio between the averaged FA in the ROI and the reference voltage. Using the circular polarization, a 3.2-fold transmission efficiency increase was measured. A much larger i.e. 5.8-fold increase was measured using the linear BC polarization. The present results indicate that a metasolenoid passive resonator can largely improve transmit efficiency in a 3T MRI system. The corresponding enhancement factors were comparable to those expected by simulations [6]. Of interest, transmission efficiency was improved differently according to the birdcage polarisation. More specifically, using the BC linear excitation led to a further reduction of the input power required to reach a given FA thereby illustrating an improved transmission efficiency. When comparing results from each polarisation, we observed a +26% improvement of the transmit efficiency with the LP which has not been previously reported in the literature. In other words, the optimized response from the resonator was obtained when the BC port was excited so as to generate a magnetic field collinear to the metasolenoid axis. As illustrated by the results obtained with the CP, the metasolenoid did not interact with the BC when the magnetic field was generated in a perpendicular plane. The metasolenoid resonator designed for the present study was able to largely improve the magnetic field transmit efficiency within a targeted region. A three-fold increase was obtained with a quadrature driven birdcage. Of interest, the transmit efficiency was further improved (six-fold) for the linear driven birdcage. Additional numerical SAR studies will be conducted in order to confirm the benefits of the metasolenoid approach in vivo.
Dmitrii TIKHONENKO (Marseille), Kaizad RUSTOMJI, Christophe VILMEN, Arnaud DURAND, Georges NOUARI, Stefan ENOCH, Redha ABDEDDAIM, Marc DUBOIS, David BENDAHAN
11:00 - 12:30
#47024 - PG332 Cylindrical Metasurface for Efficient Travelling-wave Excitation for Head Imaging at 7T.
PG332 Cylindrical Metasurface for Efficient Travelling-wave Excitation for Head Imaging at 7T.
Ultra-high-field (UHF) MRI holds great potential for brain studies, offering SNR and CNR compared to clinical (1.5 and 3T) MRI [1]. At 7T (297 MHz), the shorter wavelength (~10 cm in the human body) results in severe field inhomogeneities and standing-wave effects, limiting uniform radio frequency (RF) coverage. More than a decade ago, the traveling-wave (TW) MRI method was introduced [2]. This method avoids local transmit (Tx) coils which usually requires multiple channels and complex magnitude/phase configurations. However, TW MRI has a significant drawback of low B1+ (Tx-efficiency) [3]. Several methods have been proposed recently to improve the efficiency and homogeneity of TW MRI excitation using additional passive structures: a coaxial waveguide for brain imaging [4], high-permittivity dielectric as a bore liner [5] or a local dielectric waveguide (DW) surrounding a human head [6]. However, all these structures are bulky and causing aliasing due to water signal. In this work, we propose a thin and light cylindrical metasurface (MS) represented with a flexible PCB to improve the efficiency of TW MRI at 7T that exhibits the Tx-efficiency similar to one of a DW [6].
To achieve an equivalence between a MS and a dielectric slab (DS) (th=28 mm, ε=52), the MS unit cells were designed to mimic the DS's slow-wave factor (for the Tx field localization effect). To reach equivalence, the phase shift through the dummy parallel-plate waveguide section containing one chain of the MS unit cells (a cross of сopper strips loaded with lumped capacitors C) must match that of the same length waveguide section with the DS fraction. Numerical models of MS and DS waveguides are shown in Figure 1A,B. The optimal MS unit cell (period of 17 mm, strip width of 0.3 mm) required 3.9 pF capacitance. A full model of the cylindrical MS surrounding a human head model (Figure 2A) was built upon a previously optimized single-element design. In the practical realization, copper strips of the MS are assumed be made on two sides of a thin and flexible polyimide PCB (th=0.05 mm, ε=3.5) ending with parallel-plate printed capacitors (square side of 2.4 mm) (Figure 2B) as used in [7]. The size of the parallel-plate capacitors to replace the lumped ones was determined using a phase delay comparison in a waveguide at 297 MHz. The B1+ field was calculated for the MS loaded with the Duke voxel model [8]. A CP patch antenna similar to [6] was placed at a 140 cm distance from the top of the voxel model to provide TW excitation. The complete numerical model is presented in Figure 2C. The cylindrical MS design was optimized by varying its length, radius of the cylinder and axial displacement relative to the head model to achieve optimal B1+ homogeneity and Tx-efficiency. B1+ field homogeneity was assessed as coefficient of variation (COV) of B1+ across a 180-mm-thick transverse slab that includes the entire brain. The SAR was also calculated using the CST Legacy averaging method over 10 g of tissues. The SAR-efficiency was defined as the ratio of the mean B1+ to the SAR level. For comparison, the DW from [6] also were simulated in the same TW setup. The correspondence between the MS and the DS was evaluated using the relative slow factor (RSF) shown in Figure 1C. The best coincidence occurred at an RSF value of 0. The frequency-dependent RSF reached a minimum of 297 MHz, demonstrating that the optimized MS provided the same phase delay as the DS at the Larmor frequency of a 7T scanner. Figure 3A presents B1+ in the central sagittal plane of the model obtained using different configurations. Figure 3B shows the parametric results obtained in the presence of the proposed MS, including the B1+ homogeneity, Tx-efficiency, pSAR10g and SAR-efficiency values calculated for a fixed MS diameter of 280 mm and various lengths. Figure 4A shows numerically simulated B1+ in the central sagittal slice using the optimal configuration (410 mm length, 280 mm diameter) of the MS and the DW. As seen in the table (in Figure 3B), the MS with a length of 410 mm provides the best compromise between the COV, Tx-efficiency, pSAR10g and SAR-efficiency. As shown in Figure 4B, the proposed MS substantially improves the RF field distribution compared to the DW. Importantly, the proposed MS provides substantially better B1+ homogeneity (by 0.4%), Tx-efficiency (by 22.3%), pSAR10g (by 7.5%) and SAR-efficiency (by 27.1%) compared to the state-of-the-art DW, while also possessing advantageous features such as reduced thickness and weight improving the patient’s comfort. In this work, we numerically investigated a novel cylindrical MS for human head TW MRI at 7T with improved efficiency and homogeneity. The MS geometry is based on a grid consisting of copper strips loaded with parallel-plate capacitors printed on a flexible polyimide substrate. The proposed MS can be used in 7T MRI applications, where convenient access and high Tx-efficiency are required simultaneously (e.g., in fMRI).
Kristina POPOVA (St. Petersburg, Russia), Georgiy SOLOMAKHA, Stanislav GLYBOVSKI, Xiaotong ZHANG, Yang GAO
11:00 - 12:30
#45923 - PG333 Open waveguide loaded with a sandwich-like metamaterial for preclinical MRI.
PG333 Open waveguide loaded with a sandwich-like metamaterial for preclinical MRI.
The traveling-wave MRI (twMRI) approach using a parallel-plate waveguide (PPWG) enables imaging with a larger field of view without cutoff frequency limitations [1]. Previous studies have shown that this scheme yields images with reasonable uniformity but lower signal-to-noise ratio (SNR) values [2]. However, incorporating a metamaterial-loaded PPWG has been demonstrated to improve SNR [3]. In this work, we propose an alternative approach: a double-layer metamaterial-loaded PPWG filled with saline solution and integrated with a transceiver bio-inspired surface for preclinical imaging.
A bio-inspired surface coil was employed for RF signal transmission, as described in [1] (Fig. 1c). The metamaterial consisted of two arrays of 3 × 50 C-shaped units fabricated from copper sheets (thickness = 35 microm, s = 5.96 × 10⁷ S/m) laminated onto an FR4 dielectric substrate (e = 4.35, tan(d) = 0.008, thickness = 1 mm, dimensions = 500 mm × 40 mm), forming a sandwich-like structure (Fig. 1b). Each C-shaped unit had a diameter of 50 mm, a 3 mm gap, and a 3 mm strip width (Fig. 1a). Phantom imaging was conducted using the PPWG filled with saline solution, with the metamaterial structure inserted inside (Fig. 1d). The bio-inspired surface coil, positioned outside the waveguide and parallel to the plates, handled both RF transmission and reception. To validate this approach, images were acquired using a standard gradient-echo sequence with the following parameters: TE/TR = 4 ms/100 ms, FOV = 40 × 40 mm², matrix size = 256 × 256, flip angle = 45°, slice thickness = 1 mm, and NEX = 1. For comparison, additional phantom images were obtained using an in-house birdcage coil (length = 5 cm, diameter = 4 cm, 4 rungs). All experiments were performed on a 7T/30 cm Bruker scanner (Bruker BioSpin MRI GmbH, Germany). Phantom images were acquired both with and without the metamaterial to validate the feasibility of this approach (Fig. 2c). Additional phantom images were obtained using a birdcage coil and the twMRI setup without the metamaterial (Fig. 2c and e, respectively). From the acquired images, SNR values and uniformity profiles were computed along the yellow line indicated in Fig. 2b (see Fig. 3a). The measured SNR values were: Sandwich-metasurface: 30.4, twMRI (no metamaterial): 24.5, In-house birdcage coil (in-hBC): 32.42. The sandwich-metasurface imaging performance was comparable to that of the in-house birdcage coil—a significant result, given that remote MRI acquisition typically suffers from reduced image quality. While all uniformity profiles exhibited similar patterns, the metamaterial-enhanced setup demonstrated higher signal intensity than the standard twMRI approach. This confirms that even a non-tuned metamaterial provides a notable improvement over conventional remote imaging methods, aligning with prior findings at 3 T [4] and 15.2 T [1]. These experimental results demonstrate that a metamaterial-loaded PPWG filled with saline solution can produce high-SNR images using the tw MRI approach. Our findings show that this method outperforms conventional twMRI techniques at 7 T in a preclinical imaging setting, offering a promising alternative for high-quality remote MRI acquisition.
Sergio SOLIS-NAJERA, Jelena LAZOVIC, Saul RIVERA DE LA LUZ, Alfredo RODRIGUEZ (Mexico City, Mexico)
11:00 - 12:30
#45619 - PG334 Bio-Inspired Surface Coil with Integrated Hilbert Metasurface for Enhanced Preclinical MRI.
PG334 Bio-Inspired Surface Coil with Integrated Hilbert Metasurface for Enhanced Preclinical MRI.
Metamaterials have demonstrated the ability to enhance the performance of RF coils in clinical MRI [1]. Preclinical MRI also requires RF coils with enhanced performance, and metamaterials provide a promising alternative to achieve this goal [2-3]. We have previously reported the experimental results using a flexible metasurface wrapped around a phantom and bio-inspired surface coil [4]. In this paper, we developed a metasurface and bio-inspired coil as a single unit: a metasurface based on the Hilbert curve [5] and a bio-inspired coil [4] were integrated into a single unit for preclinical MRI applications at 7 Tesla.
The size of the metasurface must correspond to that of the bio-inspired coil and its resonant frequency. We employed the theoretical framework developed by Chen et al. [6] to calculate the frequency for the principal mode of the Hilbert metamaterial:
fm=mc/2(2^N+1)a (1)
where m represents the harmonic number, c is the speed of light, N is the order and a is the side length. Using eq. (1) with N = 4, m = 1 and a = 3 cm, we obtained the following resonant frequency, . The prototype was constructed using copper sheets (thickness = 35 microm and s = 5.96 x 10^7 S/m) laminated onto a nonconductive board (FR4: e = 4.35 and tan(d) = 0.008, thickness = 1 mm, 6.5 cm long and 3.5 cm wide). The bio-inspired coil was laminated onto one side of the substrate, while the Hilbert fractal curve was laminated to the opposite side. Tuning and matching capacitors (0–15 pF: Voltronics Co., Salisbury, MD, USA) were directly soldered onto the surface, incorporating six parallel ceramic capacitors (American Technical Ceramics, Huntington Station, NY, USA) to achieve a total capacitance of 27 pF, as illustrated in Fig. 1.a). The coil prototype was precisely matched and tuned to 50 Ω at a frequency of 299.471 MHz, corresponding to the proton frequency for 7 T. Notably, the metasurface was not tuned or matched using passive components, as its resonant frequency was sufficiently aligned with the experimental frequency required for these tests. Fig. 1 presents a photograph of both the coil prototype and the Hilbert metasurface. To assess the effectiveness of this new coil design, phantom images were captured using a standard gradient echo sequence, with parameters set to TE/TR = 4.39 ms/200 ms, FOV = 60 mm × 60 mm, matrix size = 128 × 128, flip angle = 30°, slice thickness = 1 mm, and NEX = 1. Additionally, phantom images were obtained using a bio-inspired coil of comparable dimensions without the metamaterial for comparative analysis. All MRI experiments were performed on a 7T/30 cm Bruker imager (Bruker BioSpin MRI, GmbH, Germany). The resonant frequency of the Hilbert curve, calculated using Eq. (1), aligns closely with results reported by Motovilova and Huang [7], given similar dimensions and orders. It is important to note that in our case, the Hilbert curve metamaterial is not employed as a resonator, and no passive components were used for tuning or matching purposes. Instead, the geometrical characteristics and dimensions of the metamaterial determine the resonant frequency, making it suitable for preclinical applications at 7 Tesla. Figs. 2a) and b) display phantom images acquired with the Hilbert metasurface-integrated surface coil and the coil without the metasurface, both exhibiting excellent image quality. The signal-to-noise ratio (SNR) values were calculated from these images, resulting in SNRmeta+coil = 120 and SNRbiocoil = 80. This indicates an approximate 66% improvement in SNR for the metasurface-integrated coil. Additionally, a comparison of SNR versus depth was conducted experimentally using phantom images for both coil prototypes, as shown in Fig. 2c). The Hilbert metasurface-integrated surface coil demonstrates a significant performance enhancement over the bio-inspired surface coil, particularly in terms of field uniformity, signal-to-noise ratio (SNR), and spatial selectivity. These improvements are attributed to the metasurface, which enable more efficient manipulation of the RF field distribution. These experimental results are consistent with previous data reported by our group [4], further validating the reliability and reproducibility of the metasurface-based design strategy in high-field MRI applications. The experimental imaging results indicate that the Hilbert metasurface-integrated surface coil outperforms the bio-inspired surface coil. By incorporating a metamaterial into the surface coil, we can enhance its performance without the need for additional electronic components, thereby facilitating the development of innovative coil designs. Our findings demonstrate that integrating a metasurface can significantly improve coil performance for preclinical applications in high-field environments.
Sergio SOLIS-NAJERA, Edith TELLEZ, Saul RIVERA DE LA LUZ, Jelena LAZOVIC, Alfredo RODRIGUEZ (Mexico City, Mexico)
11:00 - 12:30
#47733 - PG335 Innovative coil with Remote Deployment and Decoupling for High-Resolution Cardiac MRI at 1.5T.
PG335 Innovative coil with Remote Deployment and Decoupling for High-Resolution Cardiac MRI at 1.5T.
Standard clinical MRI is outperformed by ultra-high-field MRI in terms of image signal-to-noise ratio (SNR). Bringing this level of quality into the clinical setting could enable more accurate pathology detection, earlier diagnosis, improved therapeutic monitoring and a deeper understanding of cardiac disease mechanisms. Our goal is to achieve such image quality in clinical MRI by increasing spatial resolution, which requires a higher SNR. The performance of the receive coil is a critical determinant of SNR. Conventional surface coils often provide insufficient SNR in standard clinical MRI due to their distance from the heart and relatively large size. The design of an intracardiac receive coil that can be deployed within an intravascular catheter and achieve submillimetre resolution is presented in this study.
The aim of this study is to develop a cardiac imaging catheter coil for use in 1.5T MRI scans, either intracavitary or transesophageal (fig1). Receive-only coils were developed and connected to a dedicated interface. To maximise SNR, the loop diameter was set at 2 cm. A decoupling method developed in our lab was used to minimise nearby electronic components and reduce RF-induced heating. Miniaturised parts included 0505-sized non-magnetic capacitors (Passive Plus, USA) and 1 mm coaxial cables (ES-France), selected based on electromagnetic simulations for signal optimisation, using Advanced Design System (ADS, Keysight Technologies, USA) and QucsStudio (RAFIsoft, Germany).
To suit intracardiac and intravascular navigation, the coil was made mechanically robust and flexible to withstand cardiac motion and anatomy. It was built on a flexible polyimide PCB and bonded to a nitinol support for shape memory. Nitinol is MRI-compatible, and the adhesive was LOCTITE 4305. The total catheter length was ~100 cm. The shaft used MRI-compatible materials:
• Outer shaft: PEBAX 63D, 3.6 mm outer diameter, with embedded MRI-safe LCP (Liquid Crystal Polymer) braid, previously tested.
• Inner shaft: PEBAX 72D with embedded MRI-compatible LCP braid.
The catheter coil was tested in vitro for MRI performance on a 1.5T scanner (Aera, Siemens Healthineers, Germany).
Two experimental setups were evaluated: immersion in a saline-filled bucket (fig2.A), and insertion into a half-heart phantom with saline and 1% agarose (fig2.B). Safety testing used PRFS-based MR thermometry to assess RF-induced heating [1]. Finally, high-resolution ex vivo images were acquired in a sheep heart to validate imaging performance (fig3). The catheter coil demonstrated excellent performance, with a 13-fold higher SNR within a 2 cm diameter region compared to conventional coils. Similar results were observed in the half-heart phantom filled with agarose gel. The remote decoupling strategy proved highly effective, ensuring efficient isolation of the coil element without physical intervention. The deployment mechanism functioned reliably, enabling precise and repeatable positioning. Ex vivo acquisitions (fig3) also yielded very high SNR values with an isotropic spatial resolution of 500 µm³, approaching the image quality reported in [2] using a non-deployable surface coil of identical diameter. MR thermometry confirmed safe RF performance, with no noticeable temperature increases on or around the coil. These results demonstrate the feasibility and performance of a flexible miniature catheter coil designed for intracardiac or transesophageal imaging at 1.5T. The high-resolution ex vivo images and high SNR obtained in both phantom and biological tissue confirm the effectiveness of the coil design for localized signal reception in deep anatomical regions. The remote decoupling strategy was effective in significantly reducing the number of electronic components in the imaging zone, thereby minimising the risk of RF heating. This was confirmed by MR thermometry, which showed no measurable temperature increase, indicating that the device can operate safely under MRI conditions. The combination of flexible PCB and nitinol provided both mechanical flexibility and shape memory, allowing the coil to adapt to dynamic cardiac environments while maintaining MRI compatibility. Further improvement of the device may include adding an inflatable balloon surrounding the coil at the tip of the catheter to facilitate coil deployment in constrained regions. Such a system would allow controlled mechanical expansion of the coil, ensuring proper deployment and contact with surrounding tissues, even in tight or stiff anatomical areas. This enhancement could further improve image quality and reproducibility in future in vivo applications. This work demonstrates the successful development of a compact, remotely deployable catheter coil capable of high-resolution structural imaging at 1.5T. The next steps to finalize the prototype include integrating cable traps to suppress common mode currents, addressing occasional imaging artifacts, and advancing toward in vivo imaging studies.
Dahmane BOUDRIES (Bordeaux), Sébastien ESTORT, Gilmus Valernst MARTIAL, Manon DESCLIDES, Sylvain CAUBET, Simon LAMBERT, Marie POIRIER QUINOT, Bruno QUESSON
11:00 - 12:30
#47683 - PG336 Simultaneous EEG-fMRI at 7 T: Optimisation of artifact detection loops to mitigate Radio Frequency field interference.
PG336 Simultaneous EEG-fMRI at 7 T: Optimisation of artifact detection loops to mitigate Radio Frequency field interference.
Simultaneous EEG-fMRI at ultra-high field (e.g. 7T) offers high temporal (sub-millisecond) and spatial (sub-millimeter) resolution, enabling non-invasive exploration of human brain function at the meso-scale. However, EEG-fMRI at 7T poses significant challenges, particularly EEG signal corruption due to the MR environment. One major issue is the ballistocardiogram (BCG) artifact on the EEG signal, mainly caused by heartbeat-induced movement in the static magnetic field. One technique to reduce BCG involves adding artifact detection loops (ADLs) on top the EEG signal detection electrode cap to directly record the BCG artifact from the EEG signal[1]. While this design was successfully implemented on lower magnetic fields, their design has not been optimised for 7T and, owing to the reduced RF wavelength, may cause significant RF field disruption that can affect image quality and subject safety. Using electromagnetic field simulations, this study examines how RF fields are pertubed by ADLs, and how this is altered by ADL size and material type.
Electromagnetic field simulations were performed using a finite-difference time-domain simulation software (Sim4life 8.0,ZMT,Switzerland). A 16-leg high-pass shielded birdcage coil (diameter=305mm, length=210mm) driven in circularly polarized mode was simulated, and tuned and matched using Optenni (Optenni Lab, Finland). This coil was loaded with a digital model of a phantom (SAM phantom,ϵ_R=45.3,α:0.87S/m,ZMT,Switzerland). A model of 4 ADLs was built based on a 7T prototype cap (BrainCap-MR7FLEX, Brain Products GmbH), resulting in wire lengths of approximately 28cm corresponding to the open loop area. These wires were then transformed to obtain lengths corresponding to 50%, 60%, 70%, 80%, 100% and 120% of the open loop area. Simulations for all wire lengths were conducted for carbon wire (CW)(ϵ_R=5,α=62695S/m) and resistive polymer (ϵ_R=5,σ=5.556S/m). Loops were projected onto the surface of the phantom and displaced 7mm away from the surface in both x and y directions to maintain constant spatial relations between the loops and phantom. The cable tree, where the wires from the loops aggregate and exit the RF coil was modelled as carbon wire and maintained constant for all simulations. Figure 1(a) depicts the simulation model. A simulation with no loops (just phantom and RF coil) was run to establish the unperturbed B1+ and SAR values.
Simulated B1+ field maps and 10g mass average SAR (SAR10g) maps were normalised to 1W total input power. These were masked to the region inside the phantom, interpolated at 1mm-isotropic resolution, and exported to Matlab (R2024a,The Mathworks,Natick,MA). Maximum Intensity Projection (MIP) B1+ and SAR maps were computed in all 3 directions for each case. From each SAR10g MIP map, the MIP map of the phantom only case was subtracted to obtain a MIP difference map, to understand the full extent of the effects of the loops on SAR10g. Figure 1(b) presents the B1+ MIP maps at each loop length for carbon wire and resistive polymer loops. In the carbon wire case, interactions with the B1+ field generally increase with increasing loop length, with maxima seen at 70–80% of the original length.
In the carbon wire case, interactions with the B1+ field increased with loop length, peaking at 70-80% of the original length, before decreasing again. At these lengths, the wire creating the open loop area was approximately a quarter of the RF wavelength in air at 7T, matching resonance conditions previously reported for EEG cable bundles.[2] In contrast, minimal B1+ field perturbations were observed with higher resistance polymer ADLs, irrespective of loop length. Figure 2 and 3 show the SAR10g MIP and MIP difference maps, indicating similar trends to the B1+ maps. In the carbon wire case, resonance effects at 70-80% loop lengths led to substantial increases in SAR, whereas no such increase was seen in the polymer case due to the resistive properties of this material. As depicted in Figure 4, SAR10g values at these lengths were increased by 21-26% compared to the phantom-only case. For the polymer, SAR10g remained stable across all loop sizes, showing no resonance effects. Carbon wire ADLs were more susceptible to RF interactions than resistive polymer ADLs. Resonance effects were observed in the carbon wire loops at lengths corresponding to a quarter of the RF wavelength at 7T, consistent with previous findings in EEG cable bundles.[2] The polymer material, with its lower conductivity, exhibited minimal RF interaction, regardless of loop size. These results suggest that ADLs made from moderately resistive materials (like carbon wire) should avoid resonant lengths to limit RF field interactions. In contrast, highly resistive loops (e.g. polymer) do not exhibit significant RF interference, regardless of size. Optimizing ADLs for EEG artifact suppression while maintaining MRI data quality and safety will require considering these material and loop size effects.
Rebecca MEAGHER (London, United Kingdom), David W. CARMICHAEL, Tracy WARBRICK, Ozlem IPEK
11:00 - 12:30
#46457 - PG337 Assessment of an optical accelerometer for motion correction in supine breast MRI.
PG337 Assessment of an optical accelerometer for motion correction in supine breast MRI.
Breast MRI is typically performed in the prone position. Supine breast MRI offers better anatomical alignment with other imaging modalities and surgical planning, but compromised by respiratory motion artifacts. We previously showed that GRICS correction with a respiratory belt improves supine breast MRI [1]. With BraCoil [2], a flexible supine breast coil, accelerometers mounted on its surface outperformed the belt - likely due to better correlation with chest motion [3].
Despite its flexibility, BraCoil may still dampen motion signals. Therefore, direct chest placement of sensors is expected to improve signal fidelity. However, MEMS-based accelerometers require batteries, which introduce susceptibility artifacts and must be positioned away from the tissue of interest.
In this study, we assess the MRI compatibility of an optical accelerometer and demonstrate its usability for respiratory motion correction in supine breast MRI.
The evaluated sensor was the optical accelerometer FOSA 3660 (Optoacoustics Ltd, Or Yehuda, Israel). The MRI was Siemens Prisma 3T. Testing included safety, interference, and motion correction performance.
Safety tests assessed RF-induced heating and gradient-induced vibrations.
i) RF heating: The sensor was installed on a phantom (Fig. 1a). Four optical temperature probes were connected to a Reflex conditioner (Neoptix Canada LP). Temperature data were recorded in 3 phases: pre-sequence (20 min), during an MRI sequence (qTSE, RF Level 1, 15 min), and post-sequence (5 min). To account for potential thermalization, the pre-sequence data were fitted using Newton’s law of cooling. The difference between the predicted temperature and the actual post-sequence temperature was then calculated.
ii) Vibrations: The accelerometer was mounted on a foam support in the MRI bore (Fig. 1c). Vibrations perpendicular to its main surface were measured using a laser vibrometer in combination with a custom fixture equipped with a prism. 8 EPI sequences were applied at various frequencies [4]. The average root mean square (RMS) displacement of the accelerometer was compared to that of a plastic block of similar shape and size.
Interference testing addressed susceptibility artifacts and RF noise emissions. For the susceptibility artifacts, a GRE sequence was applied (TR = 500 ms and TE = 26.6 ms). The artifacts were compared to those induced by an MRI-compatible accelerometer [5] with a non-magnetic battery PGEB-NM651825-PCB (PowerStream, Toronto, Canada). RF noise emission was assessed using a Siemens diagnostic tool that sampled the RF spectrum received by the MRI body coil over the resonance frequency range of 123.2 ± 0.5 MHz, without RF excitation.
To evaluate motion correction performance for supine breast MRI, the sensor was positioned on the chest of a female volunteer. A T2w TSE sequence (60 slices) was run twice: once during forced, hard chest breathing, and once during abdominal breathing. Sensor signals recorded during the hard breathing were low-pass filtered (0.3 Hz) and used for the GRICS motion correction algorithm [6]. Resulting image quality was evaluated using the sharpness index [7]. Fig. 1b shows the results of the RF heating test. The probes started slightly warmer than the MRI room (by 0.1 to 1.2 °C). The estimated temperature increase of the far end of the sensor box (“side”) during the sequence was about 0.1°C and smaller for other parts.
Vibration analysis (Fig. 1d) showed a mean RMS displacement of 0.26 ± 0.13 µm, similar to that of the plastic block (0.24 ± 0.06 µm).
As shown in Fig. 2, the susceptibility artifact caused by the optical sensor was approximately 4 mm deep, compared to 32 mm artifact from a battery-powered accelerometer.
RF spectra acquired with and without the sensor showed no detectable difference, confirming the absence of emitted RF noise.
Fig. 3a displays respiratory signals recorded during the first TSE sequence, and Fig. 3b shows the resulting images. The motion correction improved image sharpness by 21.5 ± 13.1% (p < 0.001). The RF heating tests confirmed that the sensor is safe. Even assuming an initial body temperature (37 °C), it remained well below the 43 °C safety threshold (IEC 60601-1).
It exhibited no significant induced vibrations across all tested EPI sequences, reducing the likelihood of patient discomfort, mechanical failure or added noise during acquisition.
Minor susceptibility artifacts were observed. Nonetheless, the artifact size was substantially smaller than that of a battery-powered accelerometer and would likely be even smaller under less extreme imaging parameters.
Finally, the sensor was successfully applied for the GRICS motion correction for supine breast MRI. Validation on more volunteers and comparisons with other sensors are needed to further assess its performance. The optical accelerometer is safe for direct chest placement in the MRI environment, introduces minimal artifacts, and can be effectively used for GRICS motion correction.
Karyna ISAIEVA (Nancy), Diego GONZÁLEZ SOTO, Cédric LAURENT, Pauline FERRY, Freddy ODILLE, Jacques FELBLINGER
11:00 - 12:30
#47338 - PG338 Enabling Gradient Arrays Through Digital Feedback Control.
PG338 Enabling Gradient Arrays Through Digital Feedback Control.
Gradient-array coils add extra degrees of freedom over conventional gradient systems. They have already enabled simultaneous multi-slice (SMS) excitation without SAR penalty [1], region-of-interest (ROI) gradient focusing for efficiency gains [2], and lower electric-field exposure that lifts peripheral-nerve-stimulation (PNS) limits [3].
Still, developing a practical and scalable gradient power amplifier (GPA) system has been challenging. Conventional gradient amplifiers are large, expensive, and difficult to scale beyond a small number of channels. As the number of channels increases, interactions between coils become more significant, introducing coupling effects that complicate control and require more advanced strategies. Earlier work leaned on feedforward methods that compensated for coupling using known coil models, but feedforward control strongly depends on accurate modeling, making it sensitive to model inaccuracies [4,5]. Feedback control, in contrast, directly addresses coupling effects, though it traditionally relies on costly fluxgate sensors. A low-cost analog controller was also evaluated [6], but its manual tuning becomes difficult as channel count increases, leading us to focus on digital feedback in this work. In the current implementation, we use fluxgate sensors, though the system is designed to allow future use of cheaper current sensors.
We present a modular gradient controller using high-bandwidth digital feedback. With GaN transistors and continuous-time delta-sigma ADCs, the system drives coupled coils with stable current control. Our earlier test setups involved loosely connected boards and wiring, making integration difficult and raising concerns about robustness. By adopting the Eurocard format, we consolidate the system into a single enclosure with standard mechanical and electrical interfaces, resulting in a cleaner, more maintainable design suited for practical deployment and further development.
Each controller follows the IEEE 1101.1 Eurocard standard. A 3U sub-rack holds twelve modules, allowing up to 168 channels per rack.
Our four-channel module includes an Artix-7 FPGA running real-time PID control. Currents are digitized by a 24-bit, 1.5 MSPS AD4134 ADC, which removes the need for anti-alias filters and keeps latency under 10 µs. Feedback is provided by LEM fluxgate sensors.
Amplifiers, still under development, were emulated using external GaN-based PWM setups at 666 kHz. Center-aligned PWM doubles the control bandwidth, helping avoid output filters and reducing complexity.
A plastic optical fiber (POF) board links the controller to an AMD Kintex Ultrascale FPGA acting as the MRI spectrometer, which parses Pulseq data and streams gradient waveforms optically. This minimizes EMI and is suitable for future operation inside the scanner room.
Validation used four z-elements of a 40-channel custom gradient coil in a double Maxwell configuration. This setup introduces mutual coupling, allowing the controller to be tested in conditions relevant to array systems. The digital feedback controller successfully drove four mutually coupled gradient channels, maintaining stable and independent currents. A spin-echo MRI scan of a tomato was performed using our system for the readout gradient, while phase encoding and slice selection were handled by the scanner’s gradients. The resulting image and current measurements confirm that the controller operates reliably under coupling.
At higher gradient currents, we observed image noise that likely originated from EMI at the Larmor frequency, probably caused by PWM harmonics. Twisted shielded cables and amplifier shielding significantly reduced the noise, confirming its electromagnetic origin. These results underline the importance of EMI mitigation, especially in systems without conventional output filtering. The controller captures both current and drive signals at high bandwidth, making it possible to estimate the coil and amplifier response from data. The estimated model can be fed forward while feedback corrects the remaining error, allowing predictive or adaptive control. This may let the system work with cheaper sensors instead of fluxgates, without losing stability or tracking. We built a modular digital feedback controller and tested it on four mutually coupled coils. The system maintained stable currents and produced a clean spin-echo image using the array for readout. The design supports future expansion to larger arrays and offers a base for exploring model-based control and alternative sensor types.
Ege AYDIN (Ankara, Turkey), Mehmet Emin ÖZTÜRK, Manouchehr TAKRIMI, Ergin ATALAR
11:00 - 12:30
#47298 - PG339 Field-based spatial self-registration of multi-coil hardware for B0 field control.
PG339 Field-based spatial self-registration of multi-coil hardware for B0 field control.
Acquisition of robust Magnetic Resonance Imaging (MRI) data relies on a homogeneous B0 field, but magnetic susceptibility differences in vivo can create B0 distortions that lead to artifacts and signal dropout [1–3]. Multi-coil (MC) shimming systems use an array of individually-driven generic coils to homogenize the B0 field, and have been shown to outperform low-order spherical harmonic (SH) shimming in the brain [4–8].
Most MC setups are designed as temporary inserts for existing scanners, with high-quality basis maps [5, 7–9] (maps of the field produced per shim setting for each coil) acquired by a calibration process requiring multiple hours of scan and analysis time. Shim fields are calculated as a combination of calibrated basis maps for subsequent experiments. For an applied shim field to match the calculated field, it is therefore essential to either position the hardware in precisely the same location as in calibration or have knowledge of the exact position relative to the calibration scenario [5]. Degradations in B0 field control are observed even for millimeter-scale displacements (Figure 1). Our purpose is to reliably detect misplaced hardware using only a field map acquisition, ensuring optimal performance of MC inserts.
A hardware self-registration algorithm [10] in MATLAB (Mathworks, Inc., Natick, MA) co-registers two 3D field maps: an Expected Field, which is a field map acquired at the Day 0 (i.e. calibration) position, and a Measured Field, which is the same field shape measured at Day N (Figure 2). The Expected Field is mathematically created by a combination of the calibration maps, and the Measured Field is measured after the hardware is re-placed in the scanner. The algorithm calculates the x, y, and z-translation and z-rotation needed to align the two fields in an eroded ROI through co-registration. Secondary rotations about the x- and y-axes can be considered small in practice and were, thus, not considered in this proof-of-concept.
Three Expected Field shapes were tested: the shape of a selected single MC element as a baseline, a Four Lobe field generated by only one ring of coils, and a GA Field optimized by a genetic algorithm (Figure 4). The latter two were selected for further validation.
A MC array comprising 6 rows of 8 coils (diameter 70 mm, 100 turns) was arranged on a cylindrical former (OD 20.32 cm) to produce B0 field distributions (Figure 3).
Monte Carlo simulations of 5,000 recoveries of rigid transformations, were used to calculate the average norm error of localization accuracy at ten SNR levels. Simulations were done for Simulated Basis Maps – the maps generated from Biot-Savart simulations of the hardware – as well as the calibrated Experimental Basis Maps for the hardware.
The self-registration procedure was tested in the scanner by physically shifting the MC hardware a known distance. Scanner validation was performed at seven SNR levels for one hardware position, and then at one SNR level (19±1) for ten random hardware positions. The established framework enabled accurate hardware localization through application of unique field shapes (Figure 4). Localization accuracy with well below 1 mm and 1 degree errors was achieved irrespective of the applied test field shape with SNR of at least 10. In scanner validation, both Expected Fields had an average translation error below 1 mm for SNR levels above 4.6, and an average rotation error below 1 degree for SNR levels above 2.8 and 4.6 for the GA and Four Lobe fields, respectively. For all SNR levels, the Experimental Basis Maps had higher errors than Simulated Basis Maps.
Both Expected Fields were able to recover all 10 additional controlled hardware shifts within a norm translation error of under 0.44 mm, with an average error of 0.30 mm and 0.20 mm for the Four Lobe and GA Fields, respectively. All rotation errors were recovered within 0.17 degrees, with average errors of 0.13 and 0.11 for the Four Lobe and GA Fields. A field shape optimized with a genetic algorithm allows for an average localization accuracy of 0.2 mm and 0.11 degrees from a sub-1-minute B0 mapping experiment (SNR 20), allowing for improvement of B0 control and shimming potential of hardware inserts.
Simulated, Experimental, and Measured experiments showed excellent agreement across SNR levels. Both Expected Fields achieved error below 0.5 mm and 0.5 degrees for the 10 transformations – a reasonable threshold for success given the degradation curve in Figure 1 – but the GA Field had an overall lower error.
In the future, we plan to include x- and y-rotations, which cannot be accounted for by rigid transformations alone and require qualitative updates of the basis shapes, and to test the self-registration method in vivo. Here we have presented a method for field map-based hardware self-registration for MC inserts. This addresses the requirement of precise repositioning of MC hardware inserts, allowing for excellent shim capabilities of misplaced hardware.
Isabelle ZINGHINI (Vienna, Austria), Ian MACLEOD, Carlotta IANNIELLO, Sebastian THEILENBERG, Christoph JUCHEM
11:00 - 12:30
#47811 - PG340 Understanding patient grounding with experiments on a custom phantom at 55 mT.
PG340 Understanding patient grounding with experiments on a custom phantom at 55 mT.
Advancements in Very-Low Field (VLF) MRI systems have reduced the complexity of the installation, enabling the mobility of MRI equipments. However, this comes at the cost of lower Signal-to-Noise Ratio (SNR). At 50 mT and without a Faraday cage, the human body acts as an antenna coupling external electromagnetic interference (EMI) into the imaging region [2]. Recent low-field MR literature reported noise reduction via coil optimization, EMI detection and subtraction using multiple external coils, and passive methods like grounding the patient via conductive cloths or pads [1]. Software-based deep-learning approaches have also been explored to suppress EMI [3]. In this study, we examined the impact of patient grounding on EMI coupling and identify some of the factors influencing its effectiveness in VLF MRI.
We hypothesized that patient grounding can be optimized by understanding its dependencies. A custom cylindrical phantom was constructed to replicate human electrical properties [4]. The phantom consisted of a 160-cm long, 12-cm diameter polyvinyl chloride (PVC) tube, filled with water or saline (9 g/L [5]). Two non-ferromagnetic brass screws were inserted at different positions along the tube, penetrating the PVC wall to make contact with the saline.
The phantom was placed in a VLF MRI system. Noise measurements were performed using a solenoid transmit/receive coil and a Magritek Kea2 spectrometer with its «MonitorNoise» sequence. We assessed grounding effectiveness as a function of: conductivity of the medium, distance from the imaging region (grounding position) and the presence/absence of direct electrical contact.
Noise was repeatedly recorded during 60 seconds in 50-ms time windows (4 averages). Signals were concatenated, and the standard deviation (SD) was computed for comparison. SDs were first compared w/o grounding using grouding point 1 with both water and saline to isolate conductivity effects. Two main experimental conditions were then tested using saline only (Figure 1):
1.Grounding position: screws 20 cm apart, both 50 cm from the imaging region:
Config. 1: grouding point 1 only,
Config. 2: grouding point 2 only,
Config. 3: both grouding points simultaneously;
2.Insulation effects: a 110x20 cm conductive belt (σ=3,3.10^4 S⁄m) was wrapped around the tube and grounded to evaluate the effect of capacitive coupling (PVC as insulation):
Config. A: position A,
Config. B: position B,
Config. C: spiral wrap (maximized contact area). Noise SDs were computed from the real part of the signal. In the conductivity comparison, the saline-filled tube showed higher baseline noise than water, but grounding significantly dropped it, yielding a lower final level (Figure 2).
As shown in Figure 3 (a), conductive grounding reduced the noise SD: grouding point 1 reduced SD by 93.7%, versus 36.3% for grouding point 2. Simultaneous grounding through both grouding points yielded a 98.3% reduction. Capacitive coupling results (Figure 3 (b)) showed 79.4% reduction with the belt in position A, 95.2% in B, and 98.5% when spirally wrapped (C). The comparison of water vs saline filling confirms that when the entire volume is conductive, grounding is more effective, likely due to improved current flow and a more uniform electric potential distribution within the phantom.
Both conductive and capacitive grounding can significantly reduce EMI noise in an ULF MRI setup. Using conductive grounding points resulted in the highest noise suppression, with simultaneous grounding at multiple points providing the greatest improvement. Capacitive coupling via belts was most effective when spirally wrapped.
Direct contact grounding showed better efficiency when the grounding points were closer to the imaging region, likely due to the interception of external EMI effects from affecting the remaining exposed part. Conversely, capacitive coupling to ground showed better performance at longer distance from the bore and with larger contact areas. These findings suggest that both grounding strategies can be applied to reduce noise in practical MRI environments, with capacitive methods showing promise for future clinical use due to better patient comfort.
Future work will map location, contact, and area dependencies for both methods to maximize noise reduction while maintaining ease of implementation in clinical or portable systems.
Acknowledgements
This work received support from the french government under the France 2030 investment plan and French “Investissements d’Avenir” programme, as part of the Initiative d’Excellence d’Aix-Marseille Université, A*MIDEX : AMX-2023-CI-01, AMX-23-CPJ-10 and AMX-23-EQ-FO-009, as well as ANRT CIFRE 2024/1145.
Jana EL ZAHER (Marseille), Tangi ROUSSEL, Fouad FEZARI, Djamel BERRAHOU, Amira BERGÉ-LAVAL, Redha ABDEDDAIM, Marc DUBOIS, Frank KOBER
11:00 - 12:30
#47931 - PG341 A magnetic field camera to validate magnetic gradients waveforms: a proof-of concept at 0.55 T.
PG341 A magnetic field camera to validate magnetic gradients waveforms: a proof-of concept at 0.55 T.
Magnetic resonance imaging of the lung is a challenging imaging modality given the low proton density, and short T2* relaxation times. The short T2* can be compensated by using ultra-short echo time (UTE) sequences [1]. A UTE sequence with a 3D center-out radial trajectories has been developed to capture 4D (3D+time) images of the lung [2]. One of the challenges of this sequence is the ultra-short echo time and the radial center-out trajectory making this sequence highly demanding for gradient performance. These techniques often rely on non cartesian trajectories requiring a more precise gradient control where any difference between command and output can result in gradient delay artefacts (Fig.1a) and in a lack of accuracy in the derivation of the navigator. Gradient distortions are caused by non-linearities in amplifiers and variations in coil manufacturing. To tackle these issues, current advanced methods use MRI sequences along with calibrated passive phantoms [3]. Alternatively, we suggest using high-speed magnetic field cameras (MFCs) paired with algorithms that can determine the necessary adjustments from measurements to modify MR sequences and correct gradient distortions. MFCs are arrays of magnetic sensors that allow understanding complex magnetic fields by mapping the complete magnetic field vectors over a volume. MRI poses a significant challenge for MFCs due to the high-speed magnetic fields and large volumes involved. As a result, the complexity of the reading electronics increases with higher sampling rates and a larger number of sensors. Commercial MFCs [4] that utilize NMR sensors are designed to map B0 but do not provide complete field vector information. Currently, Hall-effect sensors are the most suitable type of magnetometer for MR gradients.
The magnetic camera used in this work have been presented in [5]. The MFC has an array of 7x7 integrated 3D Hall sensors (TMAG5273) (Fig.1b), covering a total area of 31.36 cm². These sensors are set to their maximum conversion rate and dynamic range of ± 266 mT. In this setup, the sensors demonstrate an rms noise of 140 µT, translating to a magnetic field resolution of 1.4 μT/√Hz. All sensors in the array are read synchronously in parallel at 7 kHz. Further details about our MFC can be found in [5]. It was then placed at the entrance to a Siemens Magnetom Free.Max (0.55 T) tunnel to characterize the gradients waveforms of a custom UTE sequence with specific a trajectory designed to enable concurrent field monitoring, sampling 25 temporal points per spoke via a field camera. Fig. 2 illustrates both the predicted gradient trajectories and the signals captured by the 49 sensors of our MFC. Given that our sensor's range is limited to 266 mT, the gradients along the B0 axis (Z-axis) are masked for our camera. Consequently, the subsequent figures focus solely on the X and Y axes. Fig. 3 presents a direct comparison between the results obtained by the central sensor of the camera and the planned gradient trajectories for the X and Y axes. To facilitate comparison, all signals are normalized between -1 and 1. This figure suggests that the measurements taken with the MFC proceeded as intended, as the measured shapes closely resemble the expected ones. The blue areas, which do not correlate with an orange area, correspond to the rewinding gradients applied by the scanner. Additionally, due to the MFC not being perfectly aligned within the MRI, each axis of the MFC detected contributions from gradients other than the “aligned” one. This misalignment is particularly noticeable in the center of the top left graph in Fig. 3. Finally, Fig. 4 provides a detailed view of the measurement of a single trapezoid (comprising 25 points acquired by the MFC) on the X-axis, in relation to the expected shape. Precisely measuring gradient errors could facilitate the implementation of impulse response function-based correction algorithms [6]. The results obtained with the MFC are promising in this regard but require further refinement of the experimental protocol and electronic development before they can be fully used for correcting sequences. For instance, enhancing the protocol will necessitate improved synchronization between the MFC and the MRI console. Precise positioning of the camera will also ensure cleaner signals. Enhancing the electronics to improve the camera's speed and precision will enable the collection of improved data. Indeed, the sequence used in this work had to be slightly slowed down to be measurable with the MFC. In the future, it will be essential to characterize the full-speed sequences directly. We successfully demonstrated in this work the ability of our MFC to measure gradients from a UTE sequence with a 3D center-out radial trajectory. The results obtained pave the way for the adjustment of such a sequence to improve the quality of the 4D images of the lung that it enables.
Thomas QUIRIN, Timothée CAUSSIN, Alexiane PASQUIER, Hugo NICOLAS, Rose-Marie DUBUISSON, Marie POIRIER-QUINOT, Joris PASCAL (Muttenz, Switzerland)
11:00 - 12:30
#45668 - PG342 Technological Innovation in MR-Guided Radiotherapy: Clinical Impact and Dosimetric Considerations of Elekta Unity MR-Linac.
PG342 Technological Innovation in MR-Guided Radiotherapy: Clinical Impact and Dosimetric Considerations of Elekta Unity MR-Linac.
Magnetic Resonance-guided radiotherapy (MRgRT) has emerged as a significant advancement in the field of radiation oncology. The Elekta Unity MR-Linac represents a fusion of high-field diagnostic MRI with a linear accelerator, enabling real-time imaging during treatment and the possibility of daily Adaptive Radiotherapy (ART). This integration allows personalized adaptation of the treatment plan to anatomical changes, improving target precision, reducing exposure to healthy tissues, and enabling safe dose escalation. This study explores the impact of Elekta Unity on treatment workflow, dosimetry in magnetic fields, and the application of real-time motion management techniques.
The Elekta Unity MR-Linac combines 1.5 Tesla MRI with a 7 MV linear accelerator. Its capability to perform daily ART includes two main workflows: "Adapt to Position" (ATP) and "Adapt to Shape" (ATS). ATP compensates for inter-fractional translational setup errors, while ATS accounts for complex changes including rotations and deformations. Treatment planning involves Magnetic Resonance Imaging (MRI), Computed Tomography (CT) for electron density mapping, and synthetic CT generation. The Particle Transport Algorithm (PTA) within the treatment planning system (TPS) models the dosimetric perturbations induced by the magnetic field on secondary electrons. To counteract these effects, optimized beam configurations and IMRT techniques with multiple fields are applied.
The study further investigates the Electron Return Effect (ERE) at tissue-air interfaces, the asymmetric dose distribution, and the implications on buildup regions. Comprehensive Motion Management (CMM) is employed to monitor intra-fractional target motion in real time, using non-invasive, non-surrogate-based tracking. The CMM system ensures beam delivery only when the target is within a predefined gating envelope. Elekta Unity significantly improves soft-tissue visualization, enabling more precise identification of tumor boundaries compared to CBCT. MRI sequences such as T2-weighted, T1-weighted, and FLAIR can be acquired in under four minutes, streamlining the adaptive workflow. Dosimetric analysis revealed that magnetic fields induce a reduction in the electron path radius, modifying dose distribution patterns, especially at heterogeneities like tissue-air interfaces.
Dose perturbations include an increased surface dose and asymmetric penumbra. These effects are mitigated by employing multiple beam directions in IMRT planning. Adaptive planning via weight and shape optimization (Method E) allows for complete fluence re-optimization and re-segmentation, ensuring precise dose delivery despite anatomical variations.
CMM enables the detection of target movements such as respiratory motion, sudden shifts, or patient non-cooperation. Clinical applications have shown its effectiveness in various treatment scenarios including SBRT for liver and lung metastases, prostate cancer, and non-cooperative patients. The system automatically interrupts radiation delivery when the target deviates from the planned position, resuming treatment once alignment is restored. MR-guided ART represents a paradigm shift from static to dynamic treatment approaches. Daily plan adaptation allows for margin reduction and dose escalation with reduced toxicity. However, integrating MRI into the radiotherapy workflow introduces new complexities, including longer session times and susceptibility to intra-fraction motion. The magnetic field's impact on electron trajectories must be considered during planning, especially near inhomogeneities.
Elekta Unity addresses these challenges by combining real-time imaging, accurate dose calculation via PTA, and motion management through CMM. The device demonstrates how MRgRT can be safely and effectively integrated into routine clinical practice, offering high precision even in complex anatomical regions. Elekta Unity enables a new standard in personalized, adaptive radiotherapy through the integration of high-quality MRI with linear accelerator technology. The system improves tumor visualization, compensates for anatomical changes, and mitigates motion-related uncertainties with real-time monitoring. Although the adaptive workflow is more time-consuming, the benefits in terms of precision, safety, and patient tolerance are significant. Future developments in automation and motion prediction will further streamline MRgRT, consolidating its role in advanced oncologic treatments.
Antonio DE SIMONE (Verona, Italy)
11:00 - 12:30
#47765 - PG343 Towards MRI-guided focused ultrasound therapies with a portable low-field scanner.
PG343 Towards MRI-guided focused ultrasound therapies with a portable low-field scanner.
MR-guided focused ultrasound (MRgFUS) is a non-invasive medical procedure that uses focused ultrasound waves, guided by magnetic resonance imaging (MRI), to treat various medical conditions. Up to date, for MRgFUS, only high-field MRI scanners are used, meaning enormous costs and associated maintenance. Furthermore, their large footprint and safety considerations make them cumbersome, and highly restrictive in access.
In this work, we present the development of a low-field MRI scanner featuring an elliptical bore, optimized for head imaging, thus overcoming the constraints of conventional lightweight cylindrical designs [1]. This system can be made compatible with focused ultrasound technologies, paving the way for affordable neuroimaging and low-field MRI-guided FUS therapy.
The main magnet follows a Halbach configuration composed of 19 rings of Nd₂Fe₁₄B cubic magnets [2]. The field-shimming unit is designed with a similar analog principle and consists of 23 rings. Once assembled, it bolts to the main magnet. Each gradient module includes four concentric elliptical lobes. Gradient coil lobes are fabricated from water-jetted copper plates, which are curved and mounted onto custom 3D-printed PLA supports. Multiple thermal sensors are installed on each lobe to monitor local temperatures and trigger an interlock circuit in case of overheating. We control the system with MaRCoS-based control system [3, 4]. The RF amplifier is based on an open design [4], whilst the GPA is a commercial 3-axis module for up to 15 A and 15 V in each [5].
All components are housed within an aluminum frame that accommodates both the magnet and associated electronics. The enclosure measures 68 × 95 × 140 cm³ (width × height × length) and is equipped with a suspension system integrated into its wheels, allowing it to traverse both smooth and uneven surfaces. The system operates independently and requires only a standard single-phase power outlet.
The ultrasound module consists of a single-element transducer of diameter 100 mm, immersed in a water-filled pool that is coupled to the subject’s head using a flexible membrane. To position the transducer, a fiberglass pole connects it to a robotic setup in the back of the scanner. To focus the US beam, a circular 3D-printed lens with a “saw-tooth” shape channels the ultrasound beam toward the intended target.
The entire ultrasound assembly is placed inside a large elliptical RF coil, with a length of 16 cm, a major diameter of 26 cm, a minor diameter of 19 cm, and 20 turns.
A 3D RARE image is acquired, with an isotropic FoV of 28cm, an image size of 120*120*60, a TR of 1200 ms, and an echo train length of 30, with a 10 ms echo spacing, for a total scan time of under 6 minutes. Then, B0 distortions are corrected [6]
To ensure effective ultrasound coupling, the phantom must be in direct contact with the ultrasound membrane. For this purpose, the phantom was mounted on a 3D-printed base, carefully aligned with the center of both the water pool and the MRI bore.
Once positioned, the pool was filled with degassed water to optimize ultrasound transmission. During the filling process, the membrane conforms around the phantom, creating a continuous and effective acoustic interface. The first results were obtained using a 3D-printed anatomical macaque phantom. This phantom was generated based on a previously acquired CT scan. The skull extracted from the CT data was 3D printed and subsequently coated with an agarose–water jelly layer, which provides clear visibility in MRI images. As seen in figure 3, we can correctly infer all relevant anatomical features. Initial images show that the combination of low-field MRI for FUS guiding is possible, since it provides enough contrast and spatial information for correct transducer positioning. However, during experimentation, one of the primary challenges identified was securing the subject’s head, as any movement can compromise the precision of both imaging and therapy. Accurate positioning within the MRI scanner will be essential for future therapeutic applications. We have developed a versatile and fully functional bimodal system. By combining MRI and focused ultrasound technologies, we have successfully achieved MRI-guided neuroimaging using a low-field system. We are currently in the testing phase using macaque subjects, focusing on refining the positioning and fixation mechanisms. Preliminary results in the context of neuromodulation applications appear promising and will be reported in future work. Current results suggest that, in the near future, it will be feasible to perform low-field MRI-guided neuromodulation therapy.
Pablo GARCÍA-CRISTÓBAL (Valencia, Spain), Eduardo PALLAS, Fernando GALVE, Teresa GUALLART-NAVAL, Marina FERNÁNDEZ-GARCÍA, José M. ALGARÍN, Lucas SWISTUNOW, Jose BORREGUERO, Ruben BOSCH, Jesús CONEJERO, Alba EROLES, Victor VEGAS, Josep RODRÍGUEZ, Noe JIMÉNEZ, Alicia CARRIÓN, Teresa Ana TORRES, Juanjo RODRÍGUEZ-GARCÍA, Daniel SANZ-MONTRULL, Jose Luís ALONSO, José M. BENLLOCH, Francisco CAMARENA, Joseba ALONSO
11:00 - 12:30
#47863 - PG344 Acquisition and denoising of electromyographic data in an MRI environment.
PG344 Acquisition and denoising of electromyographic data in an MRI environment.
We present the development of an instrumental and software solution for the acquisition and denoising of electromyography (EMG) signals during functional MRI (fMRI) data recordings. This solution is based on a dedicated acquisition channel optimized to isolate the considerable inductive noise associated with the commutation of magnetic field gradients. It is completed by a denoising tool for evaluating and subtracting this inductive artifact.
Our solution is to design a DAQ channel perfectly synchronized with the MRI system. This DAQ channel includes BIOPAC MRI-compatible electrodes and cables, two instrumentation amplifiers and a National Instruments NI-CompactDAQ 9178 box fitted with NI-9215 (16-bit analog acquisition) and NI-9401 (digital I/O & synchronization) modules. We have checked that the hardware components do not produce any RF artifacts on the MRI images.
Acquisition is controlled by a dedicated software developed under the LabVIEW® environment using the DAQmx driver. This software ensures hardware retriggering on each TR, and the generation for each of these volumes (TR) of its own acquisition clock, enabling the acquisition of a train of samples for the duration of the TR. Each signal sample is thus acquired at exactly the same time as the corresponding sample from the previous TR. Average noise is thus calculated on the basis of perfectly synchronous trains (globally over the entire run or over a sliding time window).
The average noise is then subtracted from the initial signal, and the resulting trains are concatenated to reconstruct the signal.
Resynchronization at each TR guarantees the absence of phase-shift noise (hard to filter) between all these TRs. The quality and relevance of the EMG signals was demonstrated by implementing a Go/NoGo task using response buttons equipped with isometric FSR pressure sensors, enabling the detection of very low pressures below the button release threshold.
We showed the evidence of the correlation between the FSR sensor signals and the EMG signal, even in the absence of button switching. The acquisition of EMG signals is very informative. For example, these signals can be used to accurately determine the onset of a motor response, before it is translated into movement or whether the response is inhibited and therefore undetectable using conventional tools (force transducers, response buttons, etc.).
This methods allowed to implement new features in fMRI analysis in subtile motor tasks Our solution solves the problem of cleaning up the inductive artifact by developing a specific hardware chain that isolates the induction noise from its source.
The results are highly promising and open up interesting prospects. The prospects that are opening up concern both the equipment, with optimized amplifiers or electrodes adapted to the muscles concerned, and the acquisition methods themselves: we could envisage real-time acquisition/de-noising using, for example, an on-board system that would perform on-line cleaning, as exists in audio, for example.
Bruno NAZARIAN (Marseille), Franck VIDAL, Julien SEIN, Marion ROYER D'HALLUIN, Laure SPEISER, Jennifer T. COULL, Jean-Luc ANTON
11:00 - 12:30
#46149 - PG345 Comparison of Capabilities for Image Quality Improvement and Lymph Node Metastasis Differentiation among DWI with Reverse Encoding Distortion Correction (RDC DWI), conventional DWI and FDG-PET/CT in Non-Small Cell Lung Cancer.
PG345 Comparison of Capabilities for Image Quality Improvement and Lymph Node Metastasis Differentiation among DWI with Reverse Encoding Distortion Correction (RDC DWI), conventional DWI and FDG-PET/CT in Non-Small Cell Lung Cancer.
Lung MRI has been suggested as useful for nodule characterization and lymph node metastasis diagnosis, and DWI is an integral part of MRI for lung MRI, and apparent diffusion coefficient (ADC) maps derived from DWI also useful in these settings1-3. However, a major disadvantage of DWIs is that it is considerably prone to artifacts, particularly susceptibility artifacts at tissue interfaces and image blurring due to image distortion. Reverse encoding direction (RDC) techniques with different approaches have been suggested as useful for reducing distortion artifact and improving image quality and diagnostic performance on DWI in not only neuro, but also head and neck or prostatic MRI4, 5. Currently, no major reports are not assessed the capability of DWI with RDC technique (RDC DWI) for improving image quality and influence on lymph node metastasis diagnosis in lung cancer patients. We hypothesized that DWI with RDC technique (RDC DWI) was more useful than conventional DWI (cDWI) for improving image quality and differentiation of metastatic from non-metastatic lymph nodes on lung DWI in patients with non-small cell lung cancer (NSCLC). The purpose of this study was to compare capabilities for image quality improvement and lymph node metastasis differentiation among RDC DWI, cDWI and FDG-PET/CT in NSCLC patients.
40 pathologically NSCLC patients underwent STIR FASE imaging, RDC DWI and cDWI at 1.5T system (Vantage Orian, Canon Medical Systems), FDG-PET/CT at two PET/CT systems (uMRI550: United Imaging, Shanghai, China; or Biograph mCT: Siemens Healthneers, Erlangen, Germany) with same protocols, transbronchial or mediastinal biopsies, surgical treatment, pathological examinations and follow-up examinations. RDC DWI and cDWI were obtained by spin-echo type echo-planar imaging (SE-EPI) sequence by same parameters (TR 3350ms/TE 59ms, b vale 0 and 1000s/mm2, 6 number of excitation [NEX], FOV 300450 mm, 144128 acquisition matrix, 288432 reconstruction matrix, section thickness 6mm, slice gap -1mm, voxel size 1.01.06.0 mm3) with and without RDC technique. According to pathological examination results, 69 metastatic nodes and 69 out of 354 non-metastatic nodes, which were randomly computationally selected, were assessed in this study. For quantitative image quality evaluation, absolute distortion ratios (ADRs) between each DWI and STIR images and signal-to-noise ratio (SNRs) of each selected lymph node were determined by region-of -interest (ROI) measurements. On qualitative image quality assessments, overall image quality, artifact and lesion conspicuity of each lesion and lymph node were assessed by 5-point visual scoring systems. For comparison of diagnostic performance for lymph node metastasis, apparent diffusion coefficients (ADCs) on both DWIs and maximum values of standard uptake value (SUVmaxs) of all metastatic and non-metastatic nodes were also determined by ROI measurements. To compare each quantitative image quality index between RDC DWI and cDWI, paired t-test was performed. To compare all qualitative image quality indexes between two methods, Wilcoxon's signed rank tests were performed. Then, Student’s t-tests were compared to determine differences of each ADC and SUVmax between metastatic and non-metastatic lymph nodes. To compare diagnostic performance among RDC DWI, cDWI and FDG-PET/CT, ROC analyses were performed. Finally, sensitivity (SE), specificity (SP) and accuracy (AC) were compared among all methods by McNemar’s test. Representative cases are shown in Figure 1. Figure 2 shows compared results of all quantitative indexes between two DWIs. ADR of RDC DWI was significantly smaller than that of cDWI (p=0.003). Figure 3 demonstrates compared result of each qualitative index between two DWIs. Overall image quality and artifact of RDC DWI were significantly improved as compared with those of cDWI (p<0.0001). When compared results of each ADC and SUVmax between metastatic and non-metastatic lymph nodes, there were significant differences of both ADCs and SUV max between metastatic and non-metastatic lymph nodes (p<0.0001). Figure 4 demonstrates results of ROC analysis for diagnosis of metastatic lymph node. Area under the curve (AUC) of RDC DWI was significantly larger than that of cDWI and FDG-PET/CT (p<0.05). When applied each threshold value, SP (100 [69/69] %) and AC (89.1 [123/138] %) of RDC DWI were significantly higher than those of cDWI (SP: 84.1 [58/69] %, p=0.004; AC: 79.7 [110/138] %, p=0.0002) and FDG-PET/CT (SP: 81.2 [56/69] %, p=0.0002; AC: 81.2 [112/138] %, p=0.001). RDC technique has better potentials for improving distortion, image quality and diagnosis of lymph node metastasis as compared with conventional DWI and FDG-PET/CT in NSCLC patients.
Masahiro ENDO, Kaori YAMAMOTO, Natsuka YAZAWA, Maiko SHINOHARA, Yuichiro SANO, Masato IKEDO, Ozaki MASANORI, Masao YUI, Takahiro UEDA, Masahiko NOMURA, Takeshi YOSHIKAWA, Daisuke TAKENAKA, Yoshiyuki OZAWA, Yoshiharu OHNO (Toyoake, Japan)
11:00 - 12:30
#47387 - PG346 In situ characterization of an MRI elastography device using laser vibrometry.
PG346 In situ characterization of an MRI elastography device using laser vibrometry.
Magnetic Resonance Elastography (MRE) is a non-invasive imaging technique used to quantify the mechanical properties of tissues. This method relies on generating mechanical waves in soft tissues using an actuator synchronized with a motion-encoding gradient (MEG) sequence [1]. The emergence of new actuator designs requires precise characterization of their mechanical vibrations, both outside and inside the MRI environment, to ensure the quality and reliability of MRE-derived mechanical properties measurement [2].
Laser vibrometry has become one of the preferred solutions for measuring vibrations within the MRI environment, thanks to its electromagnetic immunity and non-contact technology [3, 4].
In this study, we characterize the vibrations generated by a commercially available pneumatic passive driver (Resoundant, Inc., United States) both inside and outside the MRI room using laser vibrometry, demonstrating the feasibility of in situ, MRI-compatible vibration measurements.
Vibration measurements, perpendicular to the surface of the passive driver, were performed using a PDV-100 laser vibrometer (Polytec, Germany). For each acquisition, a velocity profile was recorded over 3 seconds at a 48 kHz sampling rate. Thirteen mirrors were distributed across the surface of the passive driver (Fig. 1a). The driver was mounted on a custom plastic measuring bench [5] designed to minimize the transmission of mechanical vibrations (Fig. 1b, c). Vibrations were generated at 20% maximum amplitude capacity using different MRE sequences: i) clinical gradient echo sequence at 60 Hz ii) SE-EPI based 3D MRE sequence at 80 and 100 Hz in a 1.5T MR scanner.
For vibration measurements outside the MRI scanner room, the laser vibrometer was aligned perpendicular to each measurement point (Fig. 1b). Velocity profiles were acquired at all points for each MRI sequence.
For vibration measurements inside the MRI scanner room, the passive driver was placed at the entry of the MRI tunnel with its main surface parallel to the static magnetic field (B0). Vertical vibrations were measured by redirecting the laser beam through a prism (Fig. 1c). Velocity profiles were acquired only at the central point due to setup limitations.
Data were processed using MATLAB (Mathworks, 2024), the velocity signals were bandpass filtered (cutoff frequencies: 5 Hz – 500 Hz), and the displacement profiles and spectra were computed using frequency domain integration [6]. The root mean square (RMS) displacement, fundamental frequency and harmonics together with their associated amplitudes were calculated. Fig. 2 shows an example of the measurements performed outside the MRI at the central point for all tested sequences. The fundamental frequency matched the input frequency across all points and sequences, with significant harmonics detected up to the third order. The displacement amplitude decreased as the frequency increased.
Fig. 3 shows the distribution of the displacement measured outside the MRI along the center line across the X-axis of the device. Consistent with the driver’s design, a bell-shaped profile is consistently observed for all applied sequences.
Concerning the measurements performed inside the MRI, Fig. 4 shows the vibrations of the central point compared to the ones measured outside the MRI for the clinical sequence at 60Hz. On average, RMS displacement inside the MRI was 7 ± 3 % higher than outside across all sequences (Fig. 4c). This difference is within the margin of measurement uncertainty inside the MRI, estimated at ±12%(studies not shown here). The fundamental frequency observed matched the input for all sequences, consistent with outside-MRI measurements. Frequency analysis confirmed that the driver vibrates at the command frequency, both inside and outside the MRI environment. However, as higher frequencies result in lower vibration amplitudes, appropriate compensation must be applied to maintain the same wave propagation. The similarity between displacement spectra inside and outside the MRI indicates that gradient-induced mechanical vibrations potentially transmitted through the pneumatic tube do not affect driver vibrations within the frequency range used for mechanical property characterization.
Frequency and displacement measurements inside the MRI were within the uncertainty limits when compared to the outside MRI results, validating the in situ MRI characterization approach.
The implementation of a scanning laser vibrometry [7] could reduce characterization time. Improvements to the current setup could enable spatial vibrations mapping inside the MRI The Resoundant driver demonstrates accurate frequency reproduction and repeatable displacement, inside and outside the MRI environment.
The developed setup demonstrates a proof of concept for in situ characterization of MRI elastography systems using laser vibrometry, enabling benchmarking closer to real conditions between devices and risk assessment of vibration effects on patient tissue.
Diego Julian GONZALEZ SOTO, Sarah MAGUIABOU FETSE (Nancy), Cédric LAURENT, Pauline FERRY, Freddy ODILLE, Jacques FELBLINGER, Pauline M. LEFEBVRE
11:00 - 12:30
#45989 - PG347 Development of Anatomically Realistic 2D MRI Phantoms of the Head and Knee for Imaging Research and Sequence Optimization.
PG347 Development of Anatomically Realistic 2D MRI Phantoms of the Head and Knee for Imaging Research and Sequence Optimization.
Magnetic Resonance Imaging (MRI) continues to evolve rapidly, driven by innovations in deep learning, quantitative mapping, and hybrid imaging approaches. Recent advances have expanded MRI’s capabilities beyond conventional anatomy imaging to include quantitative tissue profiling [1], super-resolution reconstruction [2], and even real-time therapeutic guidance [3]. As MRI systems grow increasingly complex, there is a growing need for more versatile, anatomically realistic phantoms that replicate both structural detail and tissue-specific contrast behavior to support the testing, development, and teaching of these emerging technologies [4,5].
While many existing phantoms focus on replicating quantitative relaxation times, they remain overly simplistic in structure, lacking the anatomical complexity and clinically relevant relative contrast seen in vivo [4,6], particularly in musculoskeletal imaging, where no anatomically accurate MRI phantom of the knee currently exists. To address this gap, we present the technical development and evaluation of 3D printed sectional (2D) anatomically realistic head and knee phantoms, designed to achieve both qualitative contrast realism and quantitative relaxation times which are critical for MRI technologies development testing and validation.
Our phantom development followed four main steps: (1) manual segmentation of MRI images, (2) CAD-based modeling, (3) formulation of tissue-mimicking materials (TMMs), and (4) 3D printing, filling, and MRI validation (Fig. 1).
TMM compositions were selected based on literature [7–11] to replicate contrast behavior seen in T1- and T2-weighted imaging. MRI slices (sagittal, axial, coronal) of the head and knee were sourced from our institutional image library and manually delineated by an expert radiologist. Segmentations were exported as .svg files and modeled into hollow modules using PTC-Creo.
CAD models were scaled 1:1 of average size of the specific human anatomy simulated, exported as .stl, and sliced in BambuStudio. Printing was done using 1.75 mm PETG filament on an FDM printer (parameters in Fig. 2a). Each TMM was poured into its corresponding compartment in the 3D printed hollow modules using a beaker or syringe, sealed with film, and stored at 4 °C. Imaging was performed on a 1.5 T clinical scanner (Ingenia Evolution, Philips Healthcare) with a Head-Neck coil. Phantoms were scanned using T1w, T2w and Rhow sequences to evaluate contrast, resolution, and artefact behavior (acquisition parameters in Fig 2b). Figures 3 and 4 present the qualitative MRI results of the head and knee phantoms respectively. Figure 3a shows a reference axial MRI image of the human knee, while Figures 3b-d display the corresponding phantom images acquired using the protocols outlined in Figure 2, depicting comparable anatomical regions and contrast realism.
Figure 4a presents a sagittal MRI image of the human head, included for comparison with phantom images in Figures 4b-d, which illustrate how variations in acquisition parameters, specifically pixel size affect spatial resolution. Figure 4d highlights the anatomical fidelity of the sagittal phantom, with several key structures readily identifiable. We have developed high-fidelity, anatomically realistic MRI phantoms of the head and knee, achieving relative contrast consistent with clinical expectations in T1-weighted, T2-weighted, and proton density-weighted sequences. By comparing phantom images to human MRI references, we qualitatively confirmed their ability to reproduce tissue-specific contrast patterns and recognizable anatomical features.
Beyond visual validation, the phantoms were used to demonstrate a practical use case in image quality assessment. Varying acquisition parameters, particularly pixel size, revealed visible differences in spatial resolution and artefact expression. For example, truncation artefacts (black arrowhead, Figure 4b) were more pronounced in lower-resolution images, while anatomical detail (white arrowhead, Figure 4c) improved with higher matrix acquisition. These underscore the phantom’s utility in sequence optimization and technical benchmarking.
The ability to identify multiple anatomical structures further supports the phantom’s application in education and training, providing a reusable, ethically neutral platform for hands-on MRI instruction.
While this work was able to achieve anatomical and contrast realism, quantitative relaxometric accuracy (T1 and T2 mapping) remains an ongoing objective. Future efforts will incorporate these parameters to extend the phantom’s application to quantitative imaging research, sequence standardization, and the generation of machine learning-ready datasets. This work presents anatomically and contrast-representative MRI phantoms of the head and knee, demonstrating their value in imaging protocol evaluation and radiographic training. These models lay the foundation for future development of fully quantitative, multi-purpose MRI phantoms.
Habeeb YUSUFF (Strasbourg), Thibault WILLAUME, Elodie BRETON, Guillaume BIERRY, Jean‐Philippe DILLENSEGER
11:00 - 12:30
#46475 - PG348 Numerical derivation of synthetic FLAIR images from quantitative T1 and T2 maps with suppression of CSF partial volume and subject motion effects.
PG348 Numerical derivation of synthetic FLAIR images from quantitative T1 and T2 maps with suppression of CSF partial volume and subject motion effects.
Fluid Attenuated Inversion Recovery (FLAIR) is a widely used sequence for clinical diagnosis [1]. The construction of synthetic FLAIR (synFLAIR) images from quantitative MRI (qMRI) data may suffer from CSF partial volume (PV) effects [2,3], yielding false positives. To overcome this issue, several approaches such as deep-learning-based techniques [4,5] have been proposed. Here, a numerical algorithm for synFLAIR image calculation from T1 and T2 maps with suppression of CSF PV and motion artefacts is described.
Data were acquired on a 3T scanner for 5 healthy subjects (29-79y) and 20 patients with essential hypertension (50-70y). For each subject, acquisition comprised: T1 mapping: variable flip angle method, FoV=256mm x 224mm x 160mm, 1mm isotropic resolution, FA=4/24°, TR/TE=16.4/6.7ms, including B1 and B0 mapping [6]; T2 mapping: five turbo spin echo data sets, FoV=256mm x 176mm x 125mm, resolution 1mm x 1mm x 2.5mm, TR=10s, TE={17,86,103,120,188}ms. T1 and T2 calculation was performed according to [7] and [8], respectively.
For synFLAIR image calculation, the following parameters were assumed at 3T: T1_CSF=4.5s [9], T2_CSF=2s [10], maximum tissue T2: T2_max=90ms [11,12]. The algorithm comprises the following five steps:
(A) Processing of T1 map: A mask of pure CSF is derived from T1>2/3*T1_CSF and subdivided into inner (ventricles) and outer (sulci) CSF (Fig. 1a). CSF PVs are detected according to [13], deriving an enhancement parameter (EP) from T1 gradients (Fig. 1b). A parameter V is obtained by fitting T1 versus EP, highlighting areas of potential CSF PVs (Fig. 1c). These areas are added to the outer CSF via mask growing, resulting in the final mask CSF1 (Fig. 1d). Please note that CSF1 comprises both pixels with pure CSF and with CSF PVs. Tissue R1 (R1_tiss) is derived as 1/T1 outside of CSF and extrapolated across the CSF areas indicated in CSF1. The CSF PV in the T1 map (pv1, see Fig. 1e), is then derived by assuming approximately R1=pv1*R1_CSF+(1-pv1)*R1_tiss.
(B) Processing of T2 map: T2 maps are co-registered to T1 maps. A T2-based mask CSF2 is calculated by adding to CSF1 inside a surrounding 2-pixel-layer connected areas with T2>T2_max. R2_tiss and the CSF PV in the T2 map (pv2, see Fig. 1f) are then derived in the same way as for T1. An effective T2 is calculated: T2_eff=T2 (for pv2=0) and T2_eff=1/R2_tiss (for pv2>0).
(C) Creation of background image (BI) from T1 map: A pseudo proton density (PD) map is derived from the T1 map according to [9] and a (purely T1-based) BI is calculated as BI=(1-pv1)*PD2 (Fig. 2a). Here, the first factor suppresses CSF, the second factor introduces PD-weighting and an additional pseudo-T2-weighting across white (WM) and gray (GM) matter since the quotient T2(GM)/T2(WM) matches roughly PD(GM)/PD(WM) [11,12,14].
(D) Creation of synFLAIR images: synFLAIR=BI*f(T2_eff) where f provides a T2-based signal enhancement and is given by: f=1 (for T2_eff =T2_max). For TE, 200ms is chosen.
(E) Motion correction: To suppress false positives due to motion artefacts in the T2 map, T2-based enhancement is only allowed for areas that also have elevated T1 values (average inner T1 must be at least 1% larger than the surrounding T1). Figure 2 shows for a healthy subject BI (Fig. 2a), synFLAIR (Fig. 2b) and conventional 2D FLAIR (Fig. 2c) and 3D FLAIR (Fig. 2d). synFLAIR does not show any false positives due to CSF PVs. Figure 3 shows for a patient with WM lesions (79y, arterial hypertension) four slices from synFLAIR (Fig. 3a) and from a conventional 3D FLAIR (Fig. 3b). synFLAIR highlights identical areas and is again free from false positives due to CSF PVs. Figure 4 shows the effects of motion correction: motion artefacts in the T2 map (Fig. 4a) result in false positives in the uncorrected synFLAIR (Fig. 4c). As the respective areas are not salient in the T1 map (Fig. 4b), erroneous enhancement is suppressed in the corrected synFLAIR (Fig. 4d). The presented algorithm for synFLAIR image generation from quantitative T1 and T2 maps is free from false positives due to CSF PV effects. It also includes the suppression of motion artefacts in the T2 map which are prone to cause erroneous hyper-intensity in synFLAIR. The synthetic FLAIR images presented here enhance the same areas as conventional FLAIR data. A potential improvement would be an additional correction for motion artefacts in the T1 map as these may reduce the quality of the CSF1 mask and hamper CSF suppression. The calculation of synthetic FLAIR images from T1 and T2 maps may help to save the scanning time required for conventional FLAIR acquisition in studies employing qMRI techniques. Please note that the potential of qMRI is not limited to the construction of synthetic FLAIR images, but extends to any type of weighting, offering great potential for saving scanner time and improving diagnosis.
Ulrike NOETH (Frankfurt am Main, Germany), Nenad POLOMAC, Elke HATTINGEN, Ralf DEICHMANN
11:00 - 12:30
#47913 - PG349 Assessing the impact of partial volume effect on cerebrovascular reactivity magnitude using anatomically-based simulations.
PG349 Assessing the impact of partial volume effect on cerebrovascular reactivity magnitude using anatomically-based simulations.
Cerebrovascular reactivity (CVR) measures capacity of blood vessels to respond to demand, which is reduced in several vascular diseases [1]. CVR can be measured using blood oxygen level dependent (BOLD) MRI, typically during a block-design paradigm alternating between medical air and air with a small proportion of CO2, to induce hypercapnia. As an in vivo measure, assessing the effect of potential confounders on CVR can be challenging. Previously, different processing approaches have been assessed using simulations [2]. However, such simulations and most CVR analyses do not typically accounted for the anatomical distribution of values nor partial volume effects (PVE), contamination of binarised tissue masks with different vascular responses from other adjacent tissues [3]. In this project, we used anatomically-based simulations to assess the effect of partial volume contamination on CVR analyses.
We used an openly accessible dataset (https://doi.org/10.7488/ds/3492, n=15) to determine tissue-specific ground-truth CVR magnitude and delay distributions as briefly described below [2]. For each subject, we segmented the T1-w scan in to cerebrospinal fluid (CSF), cortical and subcortical grey (SGM) and white (WM) matter. We preprocessed the best quality BOLD scan and calculated voxelwise CVR using a delay-adjusted linear regression, as previously described [4]. As a proxy for blood vessels, which tend to have low BOLD signal and higher signal variability, [5] we calculated percentage voxelwise temporal noise-to-signal ratio and thresholded at 6%, based on visual inspection of the resulting masks. For each region, we defined CVR magnitude and delay distributions using probability density functions derived from the Gaussian kernel density estimate.
We simulated CVR magnitude and delay maps by sampling from each tissue distribution using the high-resolution (0.5mm3) Multimodal Imaging-based Detailed Anatomical (MIDA) template as a reference to define the tissue type [6]. For each simulated map, we created a simulated BOLD timecourse (Fig.1) assuming a block-design end-tidal CO2 trace (2 minutes at 40 mmHg normocapnia, 3 minutes at 50 mmHg hypercapnia alternating for 12 minutes), replicating previous work [2]. For each voxel we shifted the timecourse by the simulated delay, added per-tissue signal intensity offsets based on the BOLD baseline signal and random Gaussian noise with temporal contrast-to-noise ratios matched to the relevant tissue.
To introduce partial volume effect and reflect the spatial resolution of typical BOLD-CVR acquisitions, we downsampled the resulting simulated BOLD scans to the acquisition resolution (2.5mm3) using AntsPy with trilinear interpolation to apply a non-linear transform between the MIDA and subject-specific mean BOLD [7]. We calculated CVR using delay-adjusted linear regression [4]. For each dataset we performed 10 simulations.
We visually compared the simulated maps against representative CVR magnitude maps from the reference dataset. Using Bland-Altman plots, we assessed per-tissue CVR magnitude differences between the simulated CVR map registered to the mean BOLD space and calculated CVR after downsampling the simulated BOLD timecourse. To assess the influence of partial volume effect, we plotted CVR magnitude and delay separately against the proportion of contamination from grey matter and vessels. We found simulated CVR maps were comparable to the downsampled reference (Fig.2), After downsampling, CVR magnitude tended lower relative to the simulated reference map in all tissues, but Bland-Altman plots showed no systematic bias (Fig.3). As the proportion of blood vessel and grey matter partial volume contamination increased, simulated WM CVR magnitude increased and delay shortened, with a more pronounced effect for voxels adjacent to blood vessels (Fig.4A-B). For example, in voxels contaminated by surrounding blood vessels WM CVR magnitude was 59% higher than in uncontaminated voxels. We showed simulations informed by underlying anatomy can be used to evaluate how partial volume contamination from surrounding tissues can affect CVR magnitude and delay. As well as identifying how partial volume effects may impact CVR analyses, such simulations can be used to evaluate robustness of different methods to contamination and pre/post-processing strategies to mitigate the impact [3, 8]. Future comparisons would benefit from more accurate blood vessel segmentations and should quantify how CVR is affected in voxels adjacent to CSF. We successfully implemented anatomically-based computational simulations to assess the effect of partial volume effects on CVR magnitude, helping inform interpretation of CVR analyses and may help develop more robust CVR quantification methods.
Chia-Lin WANG, Emilie SLEIGHT, Joanna M WARDLAW, Michael J THRIPPLETON, Michael S STRINGER (Edinburgh, United Kingdom)
11:00 - 12:30
#47937 - PG350 MR-EPT conductivity determination adopting three different sequences for phase mapping.
PG350 MR-EPT conductivity determination adopting three different sequences for phase mapping.
Magnetic Resonance (MR) technique, commonly used for anatomical imaging, can also assess dielectric properties (DPs) of biological tissues. DPs describe tissues response to applied electromagnetic fields (EMF) and are essential for the planning, monitoring and optimization of medical techniques based on the use of EMF on patients [1]. MR-Electric Properties Tomography (EPT) approach reconstructs the DPs of tissues at the resonance frequency of the MR scanner from the amplitude and phase of the radiofrequency transmit magnetic induction B1 [2]. Due to the invasiveness of traditional techniques for the dielectric characterization of tissues, the possibility to use MR for this aim is very promising. MR-EPT workflow is made of two steps. First, the spatial distribution of B1 in the tissues is measured through specific sequences. Secondly, tissues DPs are reconstructed using different methods [3]. Both the sequence definition and the choice of the EPT method are complex tasks to manage, being dependent on the experimental case of interest.
The aim of this work is to investigate MR-EPT sequences in the reconstruction of the conductivity of biological tissues. To this end, ex-vivo animal muscle and fat tissues were imaged, adopting three different sequences for phase mapping and exploring the differences among the three frameworks.
The MR acquisitions were done using a 3 T body coil scanner (Skyra, Siemens) for transmission and both the body coil and a head-neck 20 channels coil (Siemens) for reception. Muscle and fat ex-vivo samples were placed in a plastic beaker within the scanner. The B1 phase was measured with three sequences: a spin-echo (SE) based vendor-built sequence, called RF map, a classical spin echo, and a balanced steady state free precession (bSSFP) based vendor-built sequence, called BEAT. Raw data were acquired and processed to obtain the phase of the signal. The conductivity was derived at 128 MHz using the forward Helmholtz method implemented in MATLAB [3]. This method is straightforward to implement and physically based but, with respect to the other EPT methods, is strongly affected by noise and boundary errors.
As reference for the MR-EPT obtained results, the conductivity of both tissues at 128 MHz was measured with the open-ended probe technique [4]. The phase map was derived from the raw data for each of the three sequences, considering reception both with the head and the body coil. The RF map sequence was characterized by an acquisition time of 8.53 minutes and by a slice thickness of 5 mm which could bring, in the in vivo case, to the loss of important anatomical details smaller than 5 mm. The SE sequence required 11.8 minutes for the acquisition but the slice thickness had a more acceptable value of 2 mm. The BEAT sequence lasted, instead, only 41 seconds and provided a slice thickness of 2.5 mm, but the obtained signal was affected by banding artifacts. These artifacts must be deleted through the tuning of the sequence parameters for the specific case of interest. Therefore, a proper shimming was applied, deleting these artifacts.
Once the phase was obtained, the conductivity of muscle and fat was derived with H-EPT and some image processing procedures such as denoising and segmentation were applied. The modal value and the standard deviation among the values obtained in the pixels of the image were computed for each tissue and each sequence. The best agreement with the open-ended probe measurements was found with the SE sequence when the head coil was used as receiver, with percentage errors of 6 % and 2.50 % for muscle and fat, respectively. In the case of the RF map, errors of 13.4 % (muscle) and 0.83 % (fat) were obtained, while errors of 76.07 % (muscle) and 33.14 % (fat) were found for the bSSFP. For the body coil case, results obtained for fat were very distant from the open probe measurements probably due to the higher level of noise when this coil was used as receiver [5]. Overall, H-EPT results, considered with the associated variability, agreed with open probe measurements. The results showed that, depending on the adopted sequence for phase mapping, some differences in the conductivity reconstruction are found. The use of a SE sequence adopting the head coil as receiver provided the best results but also the longest acquisition time. Even the RF map provided good results but long acquisition times and a too big slice thickness. On the contrary, the bSSFP sequence provided a faster acquisition with good slice thickness value but worse conductivity results and the need of tuning the parameters to delete banding artifacts. This work compared three MR sequences applied using a Skyra Siemens scanner for the measurement of the B1 phase for the conductivity derivation through EPT. The findings demonstrated that, depending on the adopted sequence and coil for reception, specific operations must be considered adjusting the framework to find the best experimental compromise.
Flavia LIPORACE (Rome, Italy), Marta CAVAGNARO, Antonio NAPOLITANO
11:00 - 12:30
#47578 - PG351 Integration of steady-state diffusion MRI with Neural Posterior Estimation (NPE).
PG351 Integration of steady-state diffusion MRI with Neural Posterior Estimation (NPE).
Diffusion-weighted steady-state free precession (DW-SSFP) [1] is a diffusion imaging sequence achieving high SNR-efficiency and strong diffusion-weighting [2] (Figure 1). Previous work has demonstrated that DW-SSFP signal measurements may be highly sensitive to microstructural features [3,4,5], motivating the use of sophisticated biophysical models for DW-SSFP investigations. However, the complicated signal-forming mechanisms of DW-SSFP [6] currently limits the integration of advanced simulations with parameter estimation routines [4].
In this work, I investigate the integration of DW-SSFP with Neural Posterior Estimation (NPE), a parameter inference technique leveraging concepts from Bayesian statistics and machine learning [7-9]. Briefly, given a prior distribution of parameters, P(θ), and a forward simulation model, f(θ→S), NPE uses simulated data pairs [D,S] to train a neural network to directly estimate the posterior distribution, P(θ | S). Once trained, experimental data can be passed to the network to perform parameter inference (Figure 2).
Previous work has demonstrated the potential of NPE for diffusion-weighted spin-echo (DW-SE) investigations [10,11]. However, the DW-SSFP signal has additional dependencies on tissue relaxation properties (T1 & T2) and B1, which must be estimated for accurate diffusion modelling [6]. From the perspective of NPE, this corresponds to a different forward model per [T1,T2,B1] combination. Training a different forward model per [T1,T2,B1] combination is infeasible, requiring the adoption of an alternative method to address signal dependencies.
Here, I address signal dependences in DW-SSFP by adapting an NPE network to estimate P(θ | S,T1,T2,B1), i.e. the posterior distribution conditioned on the measured signal and known [T1,T2,B1] values (Figure 2). The proposed network successfully accounts for DW-SSFP signal dependencies, achieving >300x acceleration and excellent agreement with conventional non-linear least-squares (NLLS). Evaluations are performed using a Tensor representation of the DW-SSFP signal.
DW-SSFP data was acquired in a single post-mortem brain (Siemens 7T; 0.85 mm iso.; 120 directions), alongside complementary turbo-inversion recovery (TIR), turbo-spin echo (TSE) and actual flip angle (AFI) [12] data for T1, T2 and B1 mapping. For full details of sequence parameters and mapping techniques, see [13]. A forward model of the DW-SSFP signal incorporating a Tensor [4] was established incorporating experimental DW-SSFP acquisition parameters, T1, T2, B1, and Tensor coefficients.
NPE was implemented using the SBI toolbox (0.23.3) [14] in Python (3.12.8) as follows:
- Priors: Uniform distributions, limits = [0, 0.5] μm/ms (diagonal components) & [-0.25, 0.25] μm/ms (off-diagonal components). A classifier network was trained [15] to ensure simulated Tensors were positive semi-definite.
- Data Pairs: 6,000,000 data pairs, corresponding to 1,000,000 simulations with 5 Rician noise levels (uniform distribution, SNR limits = [2, 50]) + noise free. Each simulation was associated with an arbitrary Tensor (prior estimate), T1, T2 and B1 (uniform distributions, limits [300, 1200] ms, [20, 80] ms, and [0.2, 1.2]).
- Training: The NPE network was implemented using neural spline flows [16], with default SBI toolbox parameters. [T1,T2,B1] conditioning was achieved by appending their values to the signal during training, creating a 1D vector corresponding to [S,T1,T2,B1] (Figure 2). The network was trained on a personal laptop in ~31 hours, converging after 210 epochs.
- Evaluation: The network was evaluated using both simulated and experimental DW-SSFP data, performing comparisons with NLLS. Figure 3a-c compares estimated Tensor coefficients as a function (a) T1, (b) T2, and (c) B1 using NPE. Excellent agreement is found with ground truth (0.2% mean difference). Figure 3d compares the accuracy of parameter estimation for NPE and NLLS associated with different SNR levels, achieving similar accuracy (0.3% mean difference). NPE benefits from ~380x acceleration versus NLLS for parameter estimation.
Figure 4 compares experimental Tensor estimates using (a) NPE and (b) NLLS. Excellent agreement is found, achieving R = 0.974 for fractional anisotropy (FA) maps. NPE additionally benefits from detailed posterior distributions, with Figure 4c displaying an example distribution for a single corpus callosum voxel. I demonstrate that NPE achieves fast and accurate parameter inference from DW-SSFP data, with the proposed conditioning approach accounting for dependencies on T1, T2 and B1. Resulting parameter estimates give excellent agreement to ground-truth simulations (Figure 3) and experimental NLLS estimates (Figure 4). The implemented routine could be adapted to incorporate more sophisticated microstructural models, including Monte Carlo simulations [17]. NPE achieves fast and accurate parameter inference for DW-SSFP investigations.
Benjamin TENDLER (Oxford, United Kingdom)
11:00 - 12:30
#47552 - PG352 A new Imageless Magnetic Resonance framework for on-site Diagnosis with a simulation case study.
PG352 A new Imageless Magnetic Resonance framework for on-site Diagnosis with a simulation case study.
Diagnostic tests are key medical tools to detect or rule out specific pathologies. Magnetic Resonance Imaging (MRI) is very attractive due to its non-invasive and non-ionizing nature, but its high cost, complex infrastructure and long scanning times, keep it enclosed in hospitals and tied to long bottlenecks [1]. Low Field (LF) MRI relaxes cost and hardware specifications at the expense of image resolution, often improved by AI post-processing [2], [3]. Yet, insofar as MR is image-based, hardware and scanning time necessities remain hefty.
Whereas previous imageless attempts detaching image from MR still worked on k-space data representation [4]–[6], in this work we advocate for a more radical Imageless MR Diagnosis (IMRD), based on raw 1D MR time signals, further lowering barriers for MR use.
An IMRD solution comprises (i) the minimal structure of hardware, (ii) the optimal information encoding in MR data and (iii) optimal models for processing such data. In this work, we showcase an IMRD proof-of-concept proposing a combination of these three components with a simulated case study, aiming to detect and quantify in silico Multiple Sclerosis (MS) lesions [7], [8].
We downloaded 17 healthy phantoms with White Matter (WM), Grey Matter (GM) and Cerbrospinal Fluid (CSF) tissues from Brainweb [9], and in-silico MS lesions were simulated in each phantom [10]. Healthy and MS-affected slices were randomly selected to build a final data set of 935 brain slices, with in-silico MS in nearly 40 % of them (Figure 1).
Next, we simulated MR signals for each slice in the data set (Figure 2). The simulated acquisitions used a ZTE-like sequence [11]. Once a radial spoke is set, an initial Inversion Recovery (IR) pulse is followed by a train of RF pulses, introducing a rewind gradient at half the TR. The time for IR (∈[0.01,2] s), the Repetition Times (TRs, ∈[10,500] ms) and Flip Angles (FA ∈[10,150] º), of the MR sequence were optimized to maximize MS tissue distinguishability by minimizing the cross-correlation between tissues’ transverse magnetization signals. Literature T1 and T2 values for WM, GM, CSF and MS at 1.5 T were used [12]–[14]. Besides, we considered two minimal-hardware acquisitions: single-gradient (Figure 2 b)— with 1-dimensional spatial encoding by applying a frequency gradient in the X-axis at 0º—and gradientless (Figure 2 c)—with the pure Free Induction Decay (FID) signal. The final sequence consisted of 30 TRs and takes less than five seconds in both acquisitions. White noise at 54 dB was injected into the simulated MR signals, a reference value extracted from our portable 72 mT scanner [15], [16].
Finally, we trained 1D Convolutional Neural Networks (1D CNNs, Figure 3) [17] to estimate the volume and presence of MS lesions from each acquisition’s 1D MR signals, leaving a test set of 110 slices from two phantoms for final model evaluation. Figure 4 shows the results in the test set. The single-spoke acquisition reported an MS lesion detection AUC of 0.95, and a volume estimation accuracy of R² close to 0.8. The gradientless setup yielded an AUC of 0.8 and an R² of 0.98. The largest missed lesions were 0.06 mL and 0.26 mL for single-spoke and gradientless, respectively. The more accurate MS volume estimation with gradientless suggests that the dephasing induced by the gradient in single-spoke accelerates the decay of transversal magnetization signals (Figure 2 b), governed by T2*, losing the relevant information that is otherwise preserved in the gradientless’ signal slower decay (Figure 2 c). Nonetheless, the overestimation at zero MS volumes, (Figure 4 d), still leaves room for improvement, and real-world MS cases are likely to require additional data processing steps. IMRD relies on raw MR signals optimally encoding information registered by minimal hardware and scan time. Our in-silico case study aimed to exemplify such an IMRD framework in practice by detecting or quantifying MS tissue within a sample. Results with this in silico prototype of IMRD solution with (i) minimal or no spatial encoding, (ii) fast MR sequences below 5 seconds and (iii) data-driven models as CNNs, suggest that IMRD frameworks could successfully answer closed diagnostic questions. Yet, different scenarios—detecting other pathological events, as stroke or liver fibrosis, or measuring the spatial features of tissues as in lissencephaly—will probably require a different articulation of components. It is therefore vital to test IMRD with more clinical enquiries, ex vivo and in vivo data, which will probably imply changes in hardware, MR sequence and data processing. If successful, IMRD approaches could widen the possibilities of LF MR scanners, bringing on-site and nearly instant MR-based diagnosis.
Alba GONZÁLEZ-CEBRIÁN (València, Spain), Pablo GARCÍA-CRISTÓBAL, Fernando GALVE, Viktor VAN DER VALK, Efe ILICAK, Marius STARING, Webb ANDREW, Joseba ALONSO
11:00 - 12:30
#47592 - PG353 Assessing measurement consistency of a novel anisotropic phantom for higher order diffusion tensor MRI sequences.
PG353 Assessing measurement consistency of a novel anisotropic phantom for higher order diffusion tensor MRI sequences.
Diffusion MRI (dMRI) provides valuable insight into tissue microstructure for clinical and research applications. Traditional diffusion tensor imaging (DTI) models diffusion as a Gaussian process, which limits its accuracy in regions with complex fibre configurations (such as crossing or bifurcating tracts) [1]. Higher-order models, including diffusion kurtosis imaging (DKI) and constrained spherical deconvolution (CSD), address these limitations by capturing non-Gaussian diffusion behaviour or by resolving multiple fibre orientations within a voxel. Despite their theoretical advantages, these models are more sensitive to noise and acquisition variability, raising concerns about reproducibility [2]. Currently, there is no standardized method to perform quality assurance on dMRI data, and there are limited studies measuring the reproducibility of higher-order tensor metrics. This study aims to investigate the consistency of a novel anisotropic diffusion phantom (PreOperative Performance, Toronto, ON) for higher-order tensor dMRI protocols. While this phantom has recently demonstrated reliability for rank-2 tensor metrics across different MRI vendors, its suitability for higher-order models remains to be established [3].
The phantom consists of fixed cylindrical synthetic filaments (diameter = 2 μm) embedded in a PVA matrix, arranged into modules that emulate linear, crossing, and branching white matter structures. It was scanned 11 times across 3 days on a 3T Discovery MR750 (General Electric HealthCare, Waukesha, WI). The protocol included a conventional DTI sequence (30 directions, b=1000 s/mm²), two high angular resolution diffusion imaging (HARDI) acquisitions (60 and 90 directions, b=1300 s/mm²), and a multi-shell DKI sequence (30 directions, b=250–3000 s/mm²). Six regions of interest (ROIs) were drawn in various locations (Figure 1). Metrics were extracted using DIPY and FSL from three models: DTI (FA, MD, AD, RD), DKI (KFA, MK, AK, RK), and CSD (GFA). Reproducibility was assessed using coefficient of variation (CoV) and intraclass correlation coefficient (ICC). DTI-derived metrics showed excellent reproducibility across all ROIs (ICC > 0.9), with HARDI acquisitions slightly outperforming DTI in reducing variability (Figure 2). FA exhibited CoVs < 10% while MD, AD, and RD had CoVs < 3%.
DKI metrics displayed greater variability (Figure 3). KFA showed moderate CoV (~4–11%) but maintained strong ICCs. MK values ranged widely in CoV (10–27%), with the highest variability in ROIs featuring fibre crossings or branching. AK and RK had the least stability, with CoV exceeding 30% in certain ROIs, reflecting model sensitivity to fitting instability at high b-values.
GFA, derived from CSD, had moderate reproducibility across acquisitions (Figure 3). CoV was lowest for HARDI-60 (2.30%) and highest for DTI (3.26%). ICCs were 0.6603 (DTI), 0.8046 (HARDI-60), and 0.8507 (HARDI-90), suggesting increasing reliability with higher angular resolution (Figure 3). The phantom showed high reproducibility for DTI metrics, supporting its use in conventional tensor-based QA. HARDI protocols provided modest improvements in complex regions, aligning with research showing increased angular resolution enhances tensor fitting [2]. In contrast, DKI parameters demonstrated moderate reproducibility, with KFA being the most stable (CoV ~7.36%, ICC 0.9361). MK, AK, and RK showed increased variability (CoV 14-18%), particularly in complex fibre regions, reflecting the sensitivity of kurtosis metrics to noise and acquisition parameters. These findings suggest DKI metrics require more stringent quality assurance protocols when used for quantitative assessment.
CSD-derived GFA exhibited improved reproducibility with increased angular resolution, as evidenced by the higher ICC values for HARDI-90 (0.8507) compared to conventional DTI (0.6603). Importantly, qualitative examination of CSD fibre orientation distribution functions (Figure 4) demonstrates the phantom's ability to accurately represent complex fibre architectures that cannot be resolved using conventional DTI. Distinct peaks were observed at expected crossing locations, suggesting the phantom’s internal geometry provides sufficient angular contrast to evaluate orientation-resolving models. However, the variability in GFA and DKI metrics highlight the need for further refinement of acquisition and processing pipelines before routine use of higher-order models in QA workflows. The PreOperative Performance phantom offers a consistent and repeatable platform for assessing conventional dMRI metrics and shows promise for evaluating more advanced modelling frameworks. These findings support the use of this phantom in future multi-centre studies but also emphasize the continued need to refine acquisition protocols and modelling techniques for higher-order diffusion analyses. Future work should expand to cover inter-scanner and inter-vendor reproducibility and evaluate phantom stability over time.
Lauren STEPHENS (Hamilton, Canada), Fergal KERINS, Norm KONYER, Michael NOSEWORTHY
11:00 - 12:30
#47906 - PG354 Phantom assessment of quantitative susceptibility mapping (qsm) acquisition acceleration.
PG354 Phantom assessment of quantitative susceptibility mapping (qsm) acquisition acceleration.
Quantitative Susceptibility Mapping (QSM) is a quantitative MRI technique that maps the spatial distribution of magnetic susceptibility, having shown promise for biomarker imaging applications [1]. For QSM to reach widespread clinical use, its acquisition time must be reduced whilst maintaining accuracy and image quality. Parallel imaging (PI) techniques are extensively used to reduce clinical acquisition time. However PI must be carefully optimised as higher levels of acceleration can degrade image quality by introducing heterogenous noise and aliasing artefact [2–4]. Phantoms can provide reference values for evaluating the performance of quantitative MRI techniques [5, 6]. This study aims to assess the impact of PI acceleration, for reducing QSM acquisition times, on the accuracy and quality of QSM maps.
A Gadolinium-based phantom was constructed to provide reference susceptibility values. The phantom consisted of a 2L water-filled container with six targets containers (diameter=25mm, length=92mm), five with varying concentrations of a Gadolinium-based contrast agent (Clariscan 279.3 mg/ml, gadoteric acid, GE Healthcare), mimicking susceptibility values in-vivo (0.052, 0.104, 0.155,0.31,0.57ppm) [7] and one containing water only. Multi-echo gradient echo magnitude and phase images were acquired at 1.5 T using recommended acquisition parameters from the QSM consensus group (see figure 1B) [8]. Parallel imaging factors were varied both in-plane (R1) and through-plane (R2), with a total of five acquisitions obtained. The acquisition with PI factors R1=2 and R2=1 was taken as the “gold-standard”.
QSM maps were reconstructed using the SEPIA Toolbox in MATLAB (2024b) [9, 10]. A “two-pass masking” based approach was performed [11] using nonlinear echo-combination[12], phase unwrapping using SEGUE[13] , background field removal using PDF[14] and dipole inversion using Iterative Tikhonov [15].
Six regions of interest (ROI) were defined in the phantom targets[16]. Mean and standard deviation susceptibility were calculated for each ROI and acquisition and plotted against expected susceptibility. Agreement was assessed using linear regression to obtain the slope, intercept and coefficient of determination (R2). The modified structural similarity index measure (SSIM) for QSM, XSIM, was used to compare image quality against the gold-standard acquisition, yielding four XSIM maps [17]. Parameters for calculating the XSIM maps (K1 = 0.01, K2 = 0.001 and L=1) were obtained from Milovic et al., 2022 [18]. PI accelerated QSM maps are shown in figure 2, illustrating the qualitative differences between acquisitions. Measured versus expected susceptibility values are shown in figure 3. Vertical error bars represent ROI standard deviations while horizontal error bars represent Gadolinium concentration uncertainty of each target. Solid lines (blue) show estimates from linear regression with dotted lines showing ideal agreement. Shaded areas are bounded by 95% confidence intervals of the fit. Fit parameters are included with associated 95% confidence intervals. In-plane, through-plane XSIM maps and mean XSIM for PI factors are shown in figure 4. Quantitative susceptibility measurements (figure 3) show good linear agreement with expected values for QSM maps with increasing PI factor: (R1=2, R2=2), (R1=3, R2=1) and (R1=3, R2=2). The results show the potential to reduce acquisition times to 33% that of standard scanning while maintaining accuracy. However, quantitative measurements are impacted when using parallel imaging factors of R1=3 and R2=3, as severe artifacts are apparent. XSIM measurements demonstrate a decrease in image quality with increasing parallel imaging factor. XSIM maps are heterogeneous, highlighting the non-uniform increase in noise for increasing parallel imaging factor. This study assessed the effect of increased parallel imaging on measured susceptibility values and image quality. Good agreement was found between ROI measurements and expected values despite increasing in-plane and through-plane PI factors. However, a parallel imaging factor of 3 for both R1 and R2 yielded severely artifacted QSM maps and poor susceptibility measurements. XSIM indicates decreasing image quality with increasing PI acceleration and highlights the heterogeneity of noise introduced by PI. Further work will look at the impact of partial fourier imaging on the accuracy and image quality and develop diamagnetic targets to characterise the full spectrum of susceptibility found in vivo.
Lawrence GUARDIANO (Dublin, Ireland), Sean COURNANE, Luis LEON VINTRO, Andrea DOYLE, Alan J. STONE
11:00 - 12:30
#47979 - PG355 Water-Fat-SPIO Phantom for Validation of Model-Based Reconstruction for Joint Estimation of Multiple Quantitative Maps using Single-Shot IR Multi-Echo Radial FLASH.
PG355 Water-Fat-SPIO Phantom for Validation of Model-Based Reconstruction for Joint Estimation of Multiple Quantitative Maps using Single-Shot IR Multi-Echo Radial FLASH.
While quantitative water-specific T1, R2* and fat-fraction (FF) mapping is of great interest in liver imaging [1,2], conventional methods are typically time-intensive, since they require individual data acquisition for each map. Here, we apply a fully non-linear model to reconstruct water-specific T1, R2*, and B0 field maps directly from k-space data [3]. By evaluating results obtained from our in-house designed and manufactured water-fat-SPIO phantom, we present one step towards validating our proposed method. Here, the goal was to design and construct a quantitative MR phantom that covers all three parameter maps with good accuracy while keeping the manufacturing process simple.
When designing our water-fat-SPIO phantom, we adopted manufacturing instructions of existing protocols [4,5]. Our phantom is comprised of 18 vials (18 mL) with varying designed fat volume percentages (0, 5, 10, 20, 40 and 100%), iron concentrations (0, 50, and 150 µg/mL) and T1 water values (800 and 1500 ms). Similar to Hines et al. we chose peanut oil and super paramagnetic iron oxide (SPIO) particles (magnetite, 5 nm, Sigma-Aldrich) to modulate fat concentration and R2*. Distilled water was doped with gadobutrol (Gadovist, Bayer Vital GmbH, Germany) to modulate water-specific T1 values. For gelling, we added Agar (2.23% w/v) over heat while stirring. All vials were placed in a 1 liter cylindrical container in two sets (L1, L2). Background was filled with distilled water.
To achieve fast multi-parameter acquisition, we combine a single-shot inversion recovery (IR) sequence with a continuous multi-echo radial FLASH readout and incorporate blip gradients between echoes for improved k-space coverage [6] (Fig. 1).
To jointly estimate water-specific T1, R2*, B0, and FF maps directly from the acquired k-space data, we model the underlying physical signal and formulate parameter estimation as a non-linear inverse problem. The model explicitly accounts for water and fat specific equilibrium and steady-state signal contributions and for their effective longitudinal relaxation rates. In addition, we consider the 6-peak fat spectrum [7] and field inhomogeneity. The forward model accounts for the radial sampling pattern, coil sensitivities and Fourier transform. The optimization problem is solved iteratively using IRGNM-FISTA [8-11] implemented and performed in BART [12], utilizing Sobolev and ℓ1-wavelet regularization.
Water-fat-SPIO data was acquired on a 3T Siemens Magnetom Vida equipped with a 20 channel head coil. First, we validated the proposed model-based reconstruction on a numerical phantom (BART) (Fig. 2a), covering a wide range of ground-truth values. Comparison of ROI-averaged simulated values to the ground truth shows overall low mean differences (Fig. 2b).
In physical phantom studies, reference maps for R2*, FF, and B0 were estimated using model-based reconstruction of steady-state multi-echo data (0.8x0.8x5 mm3) [13]. Steady-state data was extracted from the last 140 excitations of the same acquisition. T1 references were estimated using single-shot IR FLASH with single-echo readout [11]. Figure 3 shows reconstructed maps from one set of vials. To assess accuracy, ROI averaged mean values of reference and the here proposed method were compared. Resulting maps of water-specific T1, R2* and FF are in excellent agreement, as indicated by Pearson correlation coefficient. Differences in B0 maps need further investigation. To estimate manufacturing accuracy, we compared designed ground truth values to values obtained from our proposed method (Fig. 4, top). Although designed fat-fraction values were achieved adequately, we noticed deviations from the designed values in both R2* and T1. While deviations in R2* can be attributed to varying fat-fractions and its know effect on measured R2* values [5], discrepancy in T1 values can only be explained by manufacturing inaccuracies at this point. To rule out acquisition errors, we additionally performed gold-standard T1 mapping [14] using IR spin-echo water-only excitation acquisitions with varying TIs (TI = 30, 280, 530, 780, 1030, 1280, 1530 ms). We can report excellent agreement between gold-standard T1 maps and our proposed method (see Fig.4, bottom). We designed and manufactured a simple water-fat-SPIO phantom used towards validating our new proposed method. In our proposed method, we combined a non-linear model-based reconstruction with radial IR multi-echo FLASH acquisition enabling joint estimation of water-specific T1, R2*, FF, and B0 field maps from a single-shot acquisition of four seconds while maintaining high accuracy. Our developed method can potentially improve patient comfort and add valuable diagnostic information while simultaneously rendering multi-parametric quantitative MRI more feasible for clinical applications.
Vitali TELEZKI (Göttingen, Germany), Daniel MACKNER, Nick SCHOLAND, Zhengguo TAN, Moritz BLUMENTHAL, Philip SCHATEN, Xiaoqing WANG, Martin UECKER
11:00 - 12:30
#46108 - PG356 Effect of patient orientation on ultimate intrinsic SNR in the torso of a realistic body model.
PG356 Effect of patient orientation on ultimate intrinsic SNR in the torso of a realistic body model.
In MRI scans using a closed-bore MRI system, patients are put into the bore either head-first or feet-first, mainly based on the anatomical region being imaged, ensuring that the target area is centered and that patient comfort is maximized. Beyond these practical considerations, the interplay between the B0 field orientation (or equivalently, patient orientation) and the RF system may introduce variations in the imaging process. In this work, we investigated the influence of patient orientation on ultimate intrinsic SNR (uiSNR)[1], which is an intrinsic feature of RF reception unaffected by receive array design.
Realistic body model: The Duke body model from the Virtual Family (IT’IS Foundation)[2] was used for electromagnetic simulations. Frequency-dependent properties[3] were assigned to different tissues in the body model. B0 strengths of 1.5 T, 3 T, 7 T, 10.5 T, and 14 T were included in the simulations. Additionally, a homogeneous body model with average tissue properties and a mirror-symmetric body model were constructed from the Duke model and assigned tissue properties at 7 T. All body models were truncated from the neck to the thighs to reduce the computational burden (Figure 1).
Basis set construction: For each simulation, an electromagnetic basis set containing 3000 random bases was generated following the method of Guerin et al.[4]. Each basis contains an electric field and its corresponding magnetic field.
uiSNR calculation: The uiSNR was calculated in each voxel using the electromagnetic basis set [5], which involved computing B1-, the negatively rotating component of the magnetic field in the plane perpendicular to B0. The uiSNR maps were computed for two opposite B0 orientations (head-first and feet-first) and compared by calculating ratio maps. Figure 2 shows the uiSNR ratio maps between the two opposite B0 orientations in the original, the homogeneous, and the mirror-symmetric Duke model at 7 T. In the homogeneous model, regions where the uiSNR increases or decreases when B0 is reversed can be observed. These regions are relatively large and mainly appear in areas where adjacent body parts are separated by air. In the original heterogeneous model, regions with increased or decreased uiSNR appear on a smaller geometrical scale. In the mirror-symmetric model, regions with increased and decreased uiSNR always appear symmetrically in pairs. In the original model, similar patterns can also be observed to some extent in regions that are roughly symmetric.
Figure 3 shows the uiSNR ratio maps of the Duke model at different B0 field strengths. From 1.5 T to 14 T, the extent of variation between the two B0 orientations increases, which can also be observed from the histogram of uiSNR ratio (Figure 4). At 14 T, the difference in uiSNR caused by the two opposite B0 orientations can be as large as 50%. Model limitations: With the random basis set used in this work, convergence of SNR towards uiSNR was only achieved in regions deeper than about 3 cm from the body surface.
Effect of B0 orientation: Differences between uiSNR maps obtained with two opposite B0 orientations are a combined result of overall shape and heterogeneous local structures. For some positions in the body, there may exist a preferred orientation of B0. As ultra-high-field MRI exploits higher field strengths, this effect may become more relevant in the future as a limiting or boosting factor of image quality.
Transmission vs. reception: Transmission and reception are intrinsically linked, representing complementary aspects of the same physical process. If a position has a preferred orientation for signal reception due to global and local geometries, the B0 orientation may offer an advantage or disadvantage for excitation. In this work, we illustrated an interesting effect that patient orientation (or equivalently, B0 orientation) may influence the uiSNR in a realistic human body model. The extent of this effect increases with B0 strength. B0 orientation is therefore proposed as an imaging factor to be considered as B0 strength increases into the ultra-high-field range.
Yuting WANG (Heidelberg, Germany), Markus MAY, Marcel GRATZ, Mark LADD, Stephan ORZADA
11:00 - 12:30
#47705 - PG357 RYAN: quality assessment tool for multicenter fMRI data of the FUNSTAR phantom.
PG357 RYAN: quality assessment tool for multicenter fMRI data of the FUNSTAR phantom.
Quality assurance (QA) is essential to ensure consistent performance of MRI scanners, especially in multicenter studies where inter-scanner variability can impact data comparability [1,2]. Using phantom-based acquisitions allows for quantitative MRI (qMRI) assessment without biological confounds, enabling effective monitoring of scanner stability and temporal drifts [3,4]. To support QA harmonization, the Italian Ministry of Health established the RIN – Neuroimaging Network [5], comprising 23 Scientific Institutes of Hospitalization and Care (IRCCSs) working on unified protocols for acquiring, processing, and sharing qMRI data. Functional MRI (fMRI), being highly sensitive to signal fluctuations, particularly benefits from reliable QA [6,2]. In this context, we present a harmonized, multivendor QA toolbox aimed at assessing intra-site stability and inter-site reproducibility through phantom data, producing automated visual and numerical reports to support reproducibility across centers.
A QA study was conducted across 16 Italian RIN centers equipped with 3T scanners from three vendors. From 2017 to 2022, centers performed monthly scans using the FUNSTAR phantom [7] and a standardized fMRI protocol. The open-source Python-based RYAN toolbox [8,9] was used to analyze key QA metrics—SNR, SFNR, signal drift, Weisskoff radius of decorrelation (RDC), and even-odd variance—across central and peripheral ROIs. Metric trends were monitored over time and statistically compared across vendors and centers. Acceptance thresholds were defined for each metric and visualized using Bland-Altman plots [10]. Spike detection was performed [11], and intraclass correlation coefficients (ICCs) were computed to assess scanner reliability [12,13]. Metrics were visualized via boxplots, barplots, and coefficient of variation (CV) analyses. SNR and SNRt showed no central vs peripheral ROI differences, but significant inter-center variability [14,15], with outliers affecting SNR results (Fig.1,Fig.2a,Fig.2b,Fig.3a,Fig.3b). SFNR had high ICCs centrally (>0.9) but lower peripherally (Fig.2c), with vendor-dependent effects [16] (Fig.1a). Signal drift varied across ROIs and vendors, with moderate ICCs (0.3–0.4), especially in peripheral regions [17,18] (Fig.1,Fig.3c). Weisskoff RDCs correlated well across 2D and 3D planes [19] (Fig.1c,Fig.4a, Fig.4b, Fig.4c); the 3D method proved useful for detecting inter-slice correlated noise [20] (Fig.2e). Even-odd variance had low variability (ICC 0.7–0.8) (Fig.2f,Fig.3f)), though vendor-related differences were noted(Fig.1a). Over five years of longitudinal tracking, SNR, SNRt, and signal drift exhibited greater variability, whereas SFNR, RDCs, and even-odd variance remained comparatively stable [21] (Fig.4d). Spike and Weisskoff plots helped assess signal stability, and most centers remained within QA thresholds with minimal exceptions. This study introduces the RIN-Neuroimaging Network’s approach to standardize fMRI data acquisition across different vendors and sites using a common QA protocol and phantom. Key metrics—SNR, SFNR, drift, RDC, and even-odd variance—were used to evaluate performance variability. Peripheral ROIs showed more instability, especially for drift, likely due to gradient heating effects [22,23]. The Weisskoff RDC 3D extension helped identify slice-related noise [20]. Even-odd variance correlated inversely with RDC, supporting its role in quick noise estimation. ICC values confirmed high reproducibility, particularly for SFNR central and even-odd metrics [13]. Most centers operated within set thresholds, supporting scanner stability and effective QA implementation. We developed the RYAN toolbox as part of a three-year QA program in the RIN-Neuroimaging Network [5]. Monthly assessments of key metrics (SNR, SFNR, drift, RDC, even-odd) provided insights into scanner performance. RYAN runs automatically on the shared FUNSTAR database and delivers detailed reports for each center, allowing vendors to be notified in case of deviations. It visualizes signal time-courses, spikes, and threshold violations, using acceptance ranges derived from 16 scanners across three vendors. Regular QA tracking helps identify hardware/software-related changes affecting scanner performance. Future developments will include enhanced SNR estimation using 32 background ROIs and a user-friendly GUI for on-site QA checks.
Antonio NAPOLITANO (Rome, Italy), Chiara PARRILLO, Luca CAIRONE, Camilla ROSSI ESPAGNET, Lorenzo FIGÀ-TALAMANCA, Anna NIGRI, Fulvia PALESI, Alberto REDOLFI, Silvia DE FRANCESCO, Laura BIAGI, Giovanni SAVINI, Michela TOSETTI, Claudia A. M. GANDINI WHEELER-KINGSHOTT
11:00 - 12:30
#47856 - PG358 Can Radiomics Capture Diffusion Behaviour? A Phantom-Based Proof of Concept.
PG358 Can Radiomics Capture Diffusion Behaviour? A Phantom-Based Proof of Concept.
Tissue microstructure plays a central role in both clinical and neuroscience research. Currently, Diffusion Tensor Imaging (DTI) is the gold standard for the non-invasive assessment of brain white matter architecture. Despite so, conventional DTI metrics may not fully exploit the textural information embedded in diffusion-weighted images. Radiomics, a quantitative imaging approach, offers a powerful framework for extracting high-dimensional features that characterize image heterogeneity and complexity [1]. As a proof of concept, in this work we demonstrate that radiomic features can describe diffusion properties using a reference phantom specifically designed to simulate microscopic diffusion anisotropy on clinical MRI systems [2, 3].
The analysed phantom presented two concentric NMR-tubes, with inner and outer diameters of 5 mm and 10 mm, respectively. The central tube was filled with a reverse hexagonal-type liquid crystal, consisting of nanometre-scale water channels embedded within a continuous matrix of detergent and hydrocarbon, while the outer one was filled with a polymer solution. To randomize the orientation of the liquid crystal’s domains the sample was initially melted and, subsequently, domains alignment along a single direction was induced by exposing the phantom to an 11.7 T magnetic field for seven days. A total of 110 phantom acquisitions were obtained at evenly spaced time intervals from which multiple diffusion maps were computed, including Fractional Anisotropy (FA), Micro-Fractional Anisotropy (µFA), Mean Diffusivity (MD_xx, MD_yy, MD_zz), mean Isotropic Diffusivity (D_iso), and mean squared normalized Anisotropy (D_Δ^2) [4]. Five regions of interest (ROIs) were segmented using a threshold-supervised algorithm: the pure liquid crystal region (LQ), the pure polymer region (POL), and an intermediate zone (Mix) that includes contributions from both. In addition, two composite regions were defined: LQmix, obtained by merging LQ and Mix; and POLmix, obtained by merging Mix and POL. After pre-processing, 98 radiomic features were extracted from each diffusion metric–ROI combination using PyRadiomics [5]. Features statistically associated with liquid crystal domains alignment were identified using Spearman’s rank correlation coefficient (ρ), with significance defined as p-value < 0.01 after Bonferroni correction for multiple comparisons. A total of 3430 feature-metric-ROI combinations were analysed. Among these, 1723 resulted to be significantly related with liquid crystal orientation. Of these, 867 features showed a strong correlation (|ρ| ≥ 0.7), while 856 demonstrated a moderate correlation (0.3 < |ρ| < 0.7). Strongly related features were predominantly derived from the LQmix region (279/867), Mix region (222/867) and LQ (202/867) pure region. In contrast, the 60% of features not related with the alignment were associated with POL and POLmix regions. Considering the MRI metrics, strongly related features were from the majority obtained from FA (24%), D_Δ^2 (20%), MD_zz (20%), followed by MD_xx (11%), MD_yy (10%), D_iso (9%), and µFA (6%). Among the pure regions, LQ was expected to exhibit the highest significant correlation with domains alignment, as it was experimentally designed to display anisotropic behaviour. Conversely, the POL region showed isotropic diffusion across all time-spaced acquisitions, as reflected by the large number of features unrelated to domains' alignment. Focusing on MRI metrics, FA and D_Δ^2 were most frequently associated with alignment. FA quantifies the degree of diffusion anisotropy at the voxel level, while D_Δ^2 describes the anisotropic component of the diffusion tensor. Focusing on MD, the alignment effect was more evident in MD_zz than in MD_yy, MD_xx, reflecting sample geometry. Lastly, µFA showed the lowest number of strongly related features as it characterizes the anisotropy of the underlying microscopic structure but not it’s organization on the voxel level. A visual comparison of the grey-level distributions between the first and final acquisitions clearly reveals a substantial variation in the FA maps: mean FA values increased from 0.11 ± 0.02—indicative of near-isotropic diffusion—to 0.87 ± 0.03, reflecting a marked enhancement in anisotropy caused by the domain’s alignment. In contrast, the µFA maps exhibited minor variation, with the mean value remaining nearly constant (from 0.97 ± 0.01 to 0.99 ± 0.00), in line with theoretical expectations. In this work, we aimed to assess the feasibility of radiomic analysis of diffusion MRI metrics using a well-characterized phantom. The results of the statistical analysis of radiomic features were largely consistent with the expected physical behaviour of the system, demonstrating that radiomics can effectively capture changes in the context of diffusion MRI.
Agnese ROBUSTELLI TEST (Pavia, Italy), Francesca BRERO, Manuel MARIANI, Alessandro LASCIALFARI, Daniel TOPGAARD
11:00 - 12:30
#47911 - PG359 Patient-specific quantitative MR twins for synthetic previews and protocol planning.
PG359 Patient-specific quantitative MR twins for synthetic previews and protocol planning.
Subject-specific quantitative maps can be obtained using conventional quantitative MRI or MR fingerprinting. Our approach uses a fast, low-resolution quantification scan in combination with an artificial neural network. Synthetic MRI contrasts are simulated using the derived quantitative maps (PD, T1, T2, T2’, D, dB0, B1) using an approach known as synthetic imaging11,12. We used the subject-specific synthetic images for the purpose of contrast previews and slice planning. In this abstract we put such synthetic MRI to the test by comparing against real scans of the same sequences using suitable metrics.
The quantification sequence was based on the prepared snapshot FLASH as proposed by Weinmüller1 et al., which we extended to whole brain coverage (2x2x4 mm3, GRAPPA6 - 6) and acquisition-time of 8 mins, while acceleration to 4 mins is possible - data not shown. The series of acquired images with PD-, T1-, T2-, T2’-, D-, dB0-, and B1+preparation is then quantified using a 3D convolution neural network trained with simulated images for same preparations using brainweb2 phantoms
All sequences were programmed in the Pulseq standard9, which then directly used in the scanner as well as the simulation made the comparisons possible. Three typical clinical protocols chosen were the following: 1. Steady-state FLASH3 (as used in Localizer scans or SWI, with TE=1.37ms, TR=3.15ms, FA=8 deg, 1.6x1.6x1.6 mm resolution, 2. MPRAGE4 with TE=3.2ms, TR=8.16ms, FA=9 deg, TI=9ms, resolution=2 x2 x4 mm, and 3. T2w-TSE5 TI=1.11ms,TR=2.53ms, FA=90 deg ,resolution 2x2x4 mm. These protocols along with quantitative scan were performed on the volunteers at a 3T MAGNETOM Cima.X Scanner (Siemens Healthineers AG, Forchheim, Germany) after well informed written consent. Quantification maps of a volunteer was obtained with Inference time of 2s in seconds in Nvidia-RTX A4000 16 Gb GPU. For fast synthetic previews we used the Phase Distribution Graph (PDG) simulation7. Simulation time of these sequence on GPU for 3D Steady-state FLASH: 732s, 2D-MPRAGE: 128 s,2D- T2w-TSE:62s, both simulated and measured data was processed with the same reconstruction pipeline. Images were windowed to reflect similar GM/WM/CSF contrast.
Since the experiment is validated with synthetic and real data of the same subject, we used structural similarity10 of equivalent 2D slices of interpolated resolution without any form of co-registration. MRI vendors offer automatic slice planning algorithms based on a 3D localizer overview scan of the scanned anatomy. In a second experiment, we evaluate the performance of the vendor’s automatic slice planning algorithm on simulated localizer images. A confidence score derived by the algorithm indicates the quality of the predicted slice planning. Figure 1-3 show the visual comparison of our synthetic images based on the 2x2x4 mm quantification, together with the real acquisition of the same sequences, i.e. steady-state FLASH (Fig 1), MPRAGE (Fig 2), T2w-TSE (Fig 3). The volumetric confidence for Brain and Orbital part of 3D volume has least uncertainty than the real measurement as shown in the Table 1. Due to trade of in acquisition time our previews suffer from low resolution and anisotropic resolution, which leads to partial volume blurring effects prevalent in both sagittal and transversal views. Apart from that, the contrast features of all sequences were replicated evidently. large deviations are visible in the fat signals of the FLASH steady state, which we traced back to T1 mismatch in the quantification. Image metrics (at the bottom of the Fig(1-3)), and volumetric confidence values reveals that synthetic images are close to real measurement, bringing us a step closer to potential preview and patient-specific process planning. We showed that within the existing framework of MR-zero8, patient-specific synthetic preview is possible with reasonable (scan, inference and simulation) time and realistic contrast. Resolution and scan time of quantification can still be optimized with further accelerations. While similar approaches based on MR fingerprinting exist11,12, this work validates the generated quantitative digital twins for fast synthetic preview of digital twin of clinical sequences. In principle, synthetic preview can also be used with any other quantification method, if the complete list of parameters are provided.
Deepak Charles CHELLAPANDIAN (Erlangen, Germany), Simon WEINMÜLLER, Fabian WAGNER, Rainer SCHNEIDER, Jonathan ENDRES, Moritz ZAISS
11:00 - 12:30
#47960 - PG360 Longitudinal Assessment of Polymer Gel Phantom Stability Monitoring Using MRI Radiomic Texture Features.
PG360 Longitudinal Assessment of Polymer Gel Phantom Stability Monitoring Using MRI Radiomic Texture Features.
Radiomics offers a non-invasive tool for assessing changes in digital images by extracting quantitative features, which can be used with other available information to assist decision-making(1). Radiomics has been used in studies such as survival and recurrence estimation for many tumours(2,3). The phantoms have been used to assess the MRI imaging chain's performance and thus have found applicability in assessing radiomic feature reproducibility and stability due to the ease of multiple imaging. The stability of radiomics features has been shown to depend on various factors, including changes within the material under study. Heterogeneous phantoms comprising various tissue-equivalent materials to provide realistic anatomy and internal tissue structures have become attractive in assessing the stability of radiomic features under various conditions. Heterogeneous phantoms comprising various tissue-equivalent materials to provide realistic anatomy and internal tissue structures have become attractive in assessing the stability of radiomic features under various conditions. Several phantoms have been proposed in the literature to assess the stability of the radiomic features, including fruits, vegetables, and 3d printed objects, among others(4–6). The selection of a phantom to use depends on the study's objective; for example, longitudinal studies require a phantom that does not change or deteriorate with time, as this will affect the radiomics feature(5). In this study, a polymer gel phantom was chosen as a possible candidate for use in MRI radiomic feature analysis. The gel phantom has the advantage of other objects being inserted inside it without losing its integrity(7). The polymer gel phantom's stability must be determined before it can be considered. The study aims to determine the stability over time of polymer gel phantom using radiomic texture features.
A gel phantom was constructed by using the (7) approach by using Carbopol-974p powder combined with distilled water, Mn(NO3)2, and NaOH, and the mixture poured into a test tube. A jig was made to have five test tubes be suspended in air as shown in figure 1. The phantom was imaged (3.0T Phillips Ingenia) bi-weekly over a period of 12 months and pulse sequence, T2w TSE was used and the acquisition parameter is shown in Table 1. A radiomic pipeline suggested by IBIS was used for feature extraction, and the PyRadiomics software of 3D Slicer was used for segmentation and feature extraction. Pre-processing normalisation was performed using a z-score on all images, and features were determined for the segmented volumes for each acquisition. For each MRI image, a total of 104 features were extracted: shape features, first-order features and second-order features. The second features are further divided into: gray level co-occurrence matrix (GLCM), gray level dependence matrix (GLDM), gray level size zone matrix (GLSZM), gray level run length matrix (GLRLM), and neighbouring gray tone difference matrix (NGTDM). Coefficient of Variance (CV) was used to determine texture features useful for further analysis and the threshold was <10%. Preliminary results indicated that the 5 texture features of GLSZM showed repeatability over time as shown in table 2. While most of the shape features provided had the best CV values i.e <10%, they provide no information about the stability of the gel phantom. The GLCM and first-order each had one feature that was repeatable on all test tubes. The CV values for GLRLM and NGTDM features for all the test tubes were greater than 10% indicating they might no be useful in this case. Preliminary results indicated that the 7 texture features of GLSZM, showed no significant difference over time, while the shape feature provided no additional information about the stability of the gel phantom. The varying stability of all first order and second order features measured might be underestimated. Since the study is ongoing, assessment of other pulse sequences results will be added. This work establishes the acceptability of polymer gel phantom as a candidate for use as a radiomic feature stable phantom, when assessment of the identified texture features. Future studies will validate the findings with other pulse sequences.
Modisenyane S MONGANE (Bloemfontein, South Africa), Sussan N ACHO, Joyce M TSOKA-GWEGWENI
11:00 - 12:30
#47633 - PG361 Evaluating the stability and mri relaxation properties of alginate for breast phantom development.
PG361 Evaluating the stability and mri relaxation properties of alginate for breast phantom development.
Alginate hydrogels present potential for application in anthropomorphic tissue-mimicking phantoms due to their tuneable physical and magnetic properties [1]. Their capability to mimic tissue-relevant relaxation times makes them well-suited for applications in quantitative magnetic resonance imaging (qMRI), where inter-site reproducibility and consistency are essential for reliable biomarker validation and early cancer detection [2]. Despite their widespread use in biomedical applications [3], the long-term physical and functional stability of alginate-based phantoms needs to be fully characterized. This study aims to quantitatively assess the temporal stability of alginate breast phantoms by monitoring changes in MRI parameters, including T₁ and T₂ relaxation times, over an initial 28-day period. This work addresses the crucial need for standardized and reproducible qMRI calibration materials, especially in breast imaging applications, where reliable relaxation times reference values are needed for quantitative lesion assessment.
The prototype phantom consisted of twelve 15 mL Falcon tubes filled with 2% (w/v) alginate in saline (1.17% NaCl). To achieve target breast tissue relaxation times (Fig. 4) [4], two doping formulations were used: 0.42 mM NiCl₂ (for T₁ modulation) and 1.14 mM MnCl₂ (for T₂ contrast) to mimic fibroglandular tissue (FGT), and 3.10 mM NiCl₂ (T₁) and 1.13 mM MnCl₂ (T₂) to mimic adipose tissue characteristics. MRI data of the phantom (Fig. 1) were acquired on days 0, 14, and 28 using spin echo sequences (T₁: TR = 500–3500 ms, TE = 7 ms; T₂: TR = 500 ms, TE = 7–350 ms) on a 3 T Siemens Prisma. A freshly prepared phantom with the same composition was scanned at each timepoint to assess formulation reliability and temporal stability. Phantoms were equilibrated at scanner room temperature (19.2–20.2 °C) for at least 24 hours prior to imaging. Relaxation times were estimated using saturation recovery (T₁) and mono-exponential decay (T₂) models, with B₁ correction applied to account for RF inhomogeneity. Phantom reliability was assessed using the coefficient of variation (CV), whilst gel stability was evaluated using independent two-sample t-tests comparing days 0 and 28. Phantom reliability was high across sessions (days 0, 14, and 28), with T₁ and T₂ measurements demonstrating strong longitudinal consistency for both FGT- and adipose-mimicking phantoms (Fig. 2), particularly in the FGT phantoms (CV = 0.28%). Regarding phantom stability, a significant increase in T₁ was observed in adipose-mimicking phantoms (p < 0.05), while no significant change was detected in the FGT phantoms (Fig. 3). T₂ values declined in both phantom types (Δ = −10.44 ± 15.68% and −9.83 ± 12.67%), though these changes were not statistically significant. The phantoms demonstrated reliability, making them suitable tools for longitudinal qMRI assessment. The T₁ increase observed in adipose-mimicking phantoms likely reflects dopant redistribution or mild oxidation [5], whereas the greater stability (Δ = 6.66 ± 4.54%) of FGT-mimicking phantoms suggests that lower NiCl₂ content may inhibit such effects. T₂ reductions may reflect gel compaction or altered water mobility during aging [6], though high variability in standard deviation limits interpretation. The stable B₁ correction and narrow temperature range suggests that observed relaxation changes likely reflect intrinsic material behaviour rather than technical-induced variability. Despite relaxation times being systematically elevated (~13–15%) relative to literature values (Fig. 4) [3], their internal consistency across phantom types supports their use in comparative or calibration-focused qMRI studies. These findings highlight the potential of alginate-based phantoms as low-cost, tuneable models for advancing qMRI, by providing a means to assess the reliability of MRI relaxation times that mimic those of breast tissue. However, the relatively short 28-day monitoring period may underestimate long-term degradation, and variability in T₂ measurements suggests that future work should investigate strategies to improve gel uniformity and chemical stability. Additionally, exploring a broader range of dopants and storage conditions could further refine phantom stability and extend applicability to a wider set of imaging contexts. This study confirms that alginate-based breast phantoms provide reproducible qMRI measurements over short timescales while revealing sensitivity to material aging, particularly in T₁ behaviour. These results underscore the need for periodic reassessment of phantom properties in longitudinal or multi-centre qMRI studies to ensure sustained measurement reliability. Continued refinement of phantom composition will be key to enhancing long-term stability for use in standardizing breast imaging protocols and quantitative biomarker development.
Klara MIŠAK (London, United Kingdom), Agnieszka SIERHEJ, Chris A. CLARK, Simon WALKER-SAMUEL, Matthew CASHMORE
11:00 - 12:30
#47872 - PG362 Evaluation of the NEXI Model of gray matter in biophysically realistic brain substrates via Monte Carlo simulations.
PG362 Evaluation of the NEXI Model of gray matter in biophysically realistic brain substrates via Monte Carlo simulations.
Evaluating and validating biophysical models is crucial for accurately characterizing tissue microstructure via diffusion MRI. Neurite Exchange Imaging (NEXI) is a two-compartment model of brain gray matter (GM) accounting for water exchange between isotropically oriented neurites and extracellular space [1-3]. NEXI is based on the analytical Kärger model of barrier-limited exchange between two Gaussian compartments. Although widely used, NEXI has not yet been evaluated in realistic numerical substrates of tortuous and beaded neurites, with varying levels of orientation dispersion and membrane permeability, which may challenge the Kärger assumptions. Indeed, previous simulation work either used the same analytical forward model for generating signals as is used for fitting the signals [1,2], or performed intra-cellular simulations in realistic substrates only [3].
Here, we assess NEXI's performance using Monte Carlo (MC) simulations of diffusion in realistic GM substrates with packed tortuous and beaded neurites.
Three substrates with orientation dispersion targets ☰ c₂ = 0.4, 0.6, and 0.8 were generated using the CATERPillar toolbox [4]. For each substrate neurites were grown in a (100 µm)³ voxel, using overlapping spheres with both beading (amplitude = 0.3 × initial radius) and tortuosity (ε = 0.4, standard deviation of the distribution of the 3D placement of the spheres) (Fig. 1). MC simulations used intra- and extracellular diffusivities of 2 and 1 µm²/ms, respectively; 0.8 µs step duration; 10⁵ random walkers; and no permeability. For the highly dispersed substrate (c₂ = 0.4), simulations were run with permeability values of 0, 10, 20, and 30 µm/s [5-7]. Each simulation was repeated 3 times for reproducibility. Diffusion-weighted signals were generated for 5 shells: 1 (12 dirs), 2 (16), 3.5 (24), 5 (30), and 7 (40) ms/µm², at diffusion times ∆=15, 26, and 38 ms, and diffusion gradient duration δ=4.5 ms to approximate the narrow pulse condition. T2 relaxation was ignored. NEXI was fitted using the Swiss Knife Toolbox (https://github.com/Mic-map/graymatter_swissknife) [1,2,8], fixing the exchange time (tex) to an arbitrarily high value in impermeable substrates. For each ∆, Diffusion Kurtosis tensor (DKI) (up to b=3 ms/µm²) and, for the impermeable substrates, the Standard Model of white matter (SMI) [9] were also fitted. Resulting substrates had neurite volume fractions f=0.57, 0.56, and 0.55, and final c₂ = 0.42, 0.64, and 0.73, matching the targets c₂ = 0.4, 0.6, and 0.8, respectively (Fig. 1).
Impermeable substrates of varying c₂: NEXI intra- (D¡,-41%) and extracellular diffusivities (De, -33%) were lower than the nominal (free) diffusivity, which is expected due to the effects of hindered diffusion in the extracellular space and along the neurites (irregularities from beading and tortuosity) (Fig. 2a). De was also lower in high-dispersion substrates as expected. The D¡ estimates were consistent with SMI estimates of intracellular diffusivity (Da) (Fig. 2c). NEXI slightly overestimated the intra-neurite volume fraction (f, +12%). Mean diffusivity (MD) and mean kurtosis (MK) showed some time-dependence in those substrates due neurite irregularities (Fig. 2b). Da from SMI (Fig. 2c) also displayed some time dependence, particularly for lower dispersion substrates.
Permeable substrates: NEXI estimated tex between 10-31 ms (Fig. 3a), matching the theoretical expected exchange times given each permeability and cylinder radius [1,10]. Other NEXI parameters remained stable as a function of permeability. MK showed more marked time-dependence, increasing with permeability, while MD time-dependence was not affected (Fig. 3b). Realistic neurite geometry yielded some time-dependence in MD, suggesting some contribution of structural disorder in this diffusion time range. SMI-derived Da scaled with 1/√∆, a hallmark of short-range disorder [11]. However, most time dependence in MK was found in the presence of permeability, consistent with the expected dominant influence of intercompartmental exchange [1,12].
Importantly, NEXI compartment diffusivity and neurite fraction estimates remained stable across permeability values, showcasing its ability to separate contributions from each compartment and their exchange. Finally, NEXI-derived exchange times aligned very well with the theoretical expectations. By generating synthetic diffusion signals under controlled microstructural conditions, we evaluated the performance of NEXI to estimate key microstructural features of the GM brain tissue. Our findings suggest that NEXI can very reliably estimate exchange time across a range of environments with different permeability, as well as dissociate other model parameters from the exchange. Future work will focus on the influence of realistic noise levels and on the inclusion of cell bodies in the substrates.
Rita OLIVEIRA (Lausanne, Switzerland), Jasmine NGUYEN-DUC, Ileana JELESCU
11:00 - 12:30
#47623 - PG363 Effect of pH and temperature changes on the 23Na TQ Signal in agarose samples.
PG363 Effect of pH and temperature changes on the 23Na TQ Signal in agarose samples.
Slow interactions between sodium nuclei and surrounding macromolecules generate a triple quantum (TQ) signal, which can serve as a biomarker for cell viability [1–3]. Agarose, a polysaccharide often used in phantom experiments, forms a solid gel after cooling, trapping water in pores ranging from sub-nanometer to hundreds of nanometers in size [4]. Low-concentration agarose induces sodium biexponential relaxation, on timescales like those in vivo, making it a suitable model system for the in vivo sodium TQ signal [5].
pH and temperature changes both alter sodium’s molecular environment. Whilst the influence of pH changes in protein-sodium interactions have been previously investigated [6], temperature and pH studies in the ubiquitously used agarose remain scarce. Hence, this study investigates the influence of pH and temperature on sodium transverse relaxation times and the TQ/SQ ratio in agarose samples.
Measurements were performed using a 9.4T preclinical MRI (Bruker Biospec 94/20) with a linear Bruker 1H/23Na volume coil. For the pH measurements, 154 mmol NaCl solutions with 12 different pH values were prepared by adding either 154 mmol NaOH or citric acid. The final pH was measured with a VOLTCRAFT PH-100 ATC device (Conrad Electronic SE, Germany). The final pH values were: 2.48, 3.62, 4.65, 5, 6.23, 8.05, 10.10, 11.27, 11.30, 11.56, 11.60, 11.89. A 2% w/v agarose phantom was prepared with the final solution. For the temperature measurements, 2% and 4% w/v agarose phantoms with 154 mmol NaCl were filled in a cylindrical flask (2.7 cm diameter, 4 cm height). The phantoms were heated using a flexible animal heat bath around the area for uniform heating, with temperature continuously monitored via a fiber-optic sensor in the center of the phantom. The signal was acquired after stabilizing the heat bath, with no temperature change exceeding 0.2°C within 5 minutes. Temperature was raised in 5 steps from 19°C to 43°C, then the phantom was allowed to cool and two subsequent measurements were performed. A TQ time-proportional phase increment (TQTPPI) pulse sequence was used to simultaneously measure SQ and TQ signals (Fiqure 1). The TQTPPI FID was non-linearly fitted using [5]:
$$ Y(t) = \sin(\omega t + \phi_1) \cdot (A_{SQ,1}e^{-t/T_{2f}+A_{SQ,2}e^{-t/T_{2s}})+A_{TQ}\sin(3\omega t + \phi_2)\cdot (e^{-t/T_{2s}}-e^{-t/T_{2f}}) + DC, (1)$$
where $Y(t)$ is the TQTPPI FID amplitude, $A_{SQ,i}/A_{TQ}$ are the SQ and TQ amplitudes, respectively. $T_{2f}$ and $T_{2s}$ are the corresponding fast and slow transverse relaxation times. Sequence parameters were: $T_R = 400$ms, $\delta t_{evo}=200$us and $t_{mix}=125-135$us with a phase increment of 45° and 520 phase steps. Fig. 2 shows the TQ/SQ ratio versus pH (left) and T2 relaxation times (right), with error bars representing the 95% confidence interval. The TQ/SQ ratio increased slightly at pH 5, plateaued at ~19% between pH 5 and 9, and rised steeply above 50% at higher pH. After a pH of 11.60 the curve started to flatten, suggesting sigmoidal behavior.
The relaxation times showed an inverse trend. The slow component plateaued between 41–44 ms up to pH 10, while the fast component decreased from 16 to 9 ms by pH 10. Both dropped sharply after pH 10, reaching 0.37 ms and 20 ms at pH 11.89.
Fig. 3 depicts the temperature dependence of the TQ/SQ ratio and relaxation rates. Both relaxation times correlate positively with temperature, confirmed by R²adj = 0.99 for all fits. The TQ/SQ ratio decreased similarly in both samples with temperature. After cooling, it was systematically lower than expected, unlike the relaxation times, which remained unchanged. The behaviour of the TQ/SQ ratio in fig. 2 is driven by hydrogen bonding between hydroxyl groups [4]. At low pH, abundant +H ions inhibit bond formation, reducing negatively charged groups for ²³Na interaction and lowering the TQ/SQ ratio. Reduced bonding may also result in larger pores, decreasing the NaCl interaction surface and further lowering the ratio. At higher pH, more bonding enhances ²³Na-macromolecule interaction, raising the TQ/SQ ratio.
The decreased relaxation times align with stronger bonding, which increases ²³Na interaction.
For the temperature data, changes in relaxation times were likely caused by increased molecular motion’s influence on quadrupole interactions [7], not agarose structural changes, as its melting point is 65 °C.
The systematic drop in TQ/SQ ratio after cooling, despite stable relaxation times, suggests possible sample changes detectable by this ratio, requiring further investigation, especially in protein samples. The sodium TQ/SQ ratio exhibited strong pH dependence for values above 9, while remaining stable at physiological pH. Additionally, the study found a linear relationship between temperature and both relaxation times, as well as the TQ/SQ ratio. Both results confirm the sensitivity of the TQ signal on its environment and using the TQ/SQ ratio on is able to quantify this dependence.
Dominik ZEHENDER (Heidelberg, Germany), Valentin JOST, Frank ZÖLLNER, Lothar SCHAD
|
Poster hall |
12:30 |
"Friday 10 October"
Added to your list of favorites
Deleted from your list of favorites
A23
12:30 - 14:00
LUNCH SYMPOSIUM
|
Auditorium 900 |
|
LUNCH BREAK & LUNCH SYMPOSIUM
|
14:00 |
"Friday 10 October"
Added to your list of favorites
Deleted from your list of favorites
A24
14:00 - 15:30
ET1-4 - Impactful research publishing
ET1: Cycle of Research
14:00 - 15:30
Factors to consider when selecting a journal.
Jonathan MCNULTY (Keynote Speaker, Ireland)
14:00 - 15:30
My top tips in writing a high quality 'clinical' manuscript.
Paola CLAUSER (Keynote Speaker, Vienna, Austria)
14:00 - 15:30
My top tips in writing a high quality 'methods' manuscript.
David NORRIS (Prof) (Keynote Speaker, Nijmegen, The Netherlands)
|
Auditorium 900 |
|
"Friday 10 October"
Added to your list of favorites
Deleted from your list of favorites
B24
14:00 - 15:30
FT3-2 - Quantitative MRI and its confoundings
FT3: Cycle of Quality
14:00 - 15:30
Quality assessment of quantitative MRI.
Sebastian WEINGÄRTNER (Keynote Speaker, Delft, The Netherlands)
14:00 - 15:30
Quantitative MRI and its confoundings.
Siawoosh MOHAMMADI (Head of Microstructure MRI Group) (Keynote Speaker, Lübeck, Germany)
14:00 - 15:30
Use of quantitative MRI in a clinical populations.
Amy MCDOWELL (Keynote Speaker, London, United Kingdom)
|
Espace Vieux-Port |
|
"Friday 10 October"
Added to your list of favorites
Deleted from your list of favorites
C24
14:00 - 15:30
FT2 LT - Translational MRI
Brain microstructure & Function
14:00 - 14:02
#45824 - PG151 MouseFlow: a pipeline for mouse brain diffusion MRI data, generating tractograms and diffusion metrics with quality controls.
PG151 MouseFlow: a pipeline for mouse brain diffusion MRI data, generating tractograms and diffusion metrics with quality controls.
Diffusion magnetic resonance imaging (dMRI) experiments in rodents offer in-depth insight into the organisation of brain networks and their biological structure. By combining dMRI with complementary approaches such as light sheet microscopy [1] or optical coherence tomography [2], it is possible to create links between biological and functional levels of analysis for translational studies in neuroscience. However, despite the standardisation of dMRI protocols in human research, image processing and data quality assessment in rodents remain heterogeneous. The absence of unified and adapted standards for the processing stages - spatial alignment, sequence-dependent correction, and management of confounding factors - poses challenges for the reproducibility of results. Differences in acquisition parameters accentuate this variability. To address these gaps, we present MouseFlow, an open-source pipeline that standardizes the pre-processing of rodent dMRI data inspired by Tractoflow [3] and VersaFlow [4] and including the Allen Mouse Brain Atlas (AMBA) [5] to extract specific diffusion metrics and bundles based on regions of interest.
MouseFlow only needs dMRI data, including b-values and b-vectors. Table 1 outlines the tested parameters to evaluate the nine-step workflow illustrated in Fig. 1A. It uses Nextflow DSL2 [6] for ease of use, flexibility across profiles, parallelization, and container compatibility. A first recommended quality control step allows validating the data before running MouseFlow (Figure 1.B). MouseFlow pre-processing involves denoising, eddy currents correction, brain mask extraction, N4 bias correction, DTI [7] and fODF [8] reconstructions, and extraction of their respective metrics steps (Figure 1.C). The registration of the AMBA, allows extracting specific anatomical Regions of Interest (ROIs) and obtaining the seeding and tracking maps to reconstruct a whole-brain tractogram [8]. All parameters across the steps can be modified in a JSON configuration file. Depending on the user's input data, different pipeline profiles will autoselect optimised parameters. MouseFlow provides classical diffusion metrics like the fractional anisotropy (FA) or mean diffusivity (MD) (Figure 2A) as well as advanced fODF-derived measures for each ROI. MouseFlow also includes a tractography module: a seeding mask extracted from AMBA fibre tract ROIs, together with a tracking mask, enables the generation of a whole-brain tractogram (Figure 2B). Specific bundles can then be extracted from this tractogram (Figure 2C), offering detailed insights into white matter organisation. To validate the anatomical accuracy of the extracted tracts, we compared them with ground truth connectivity data from the AMBA (Figure 3). We focused on streamlines passing through the fornix and hippocampal commissure, which encompass the medial septal complex (MS). We selected experiment #100141597 from the AMBA, corresponding to an injection in the left MS, and imported the associated projection map into the subject’s native space. This map was then binarised using our tool m2m [9]. The extracted streamlines were compared to this binarised projection, revealing a strong spatial overlap. As shown in Figure 3, the streamlines intersecting the MS are fully contained within the AMBA connectivity map, indicating that the tracts derived using MouseFlow closely match the expected anatomical projections. We provide a robust mouse brain dMRI processing and analysis pipeline, specifically designed to meet the unique challenges of dMRI in mice. In preclinical imaging, it is essential to have homogeneous and transparent processing tools to facilitate collaboration between different laboratories. Withith this in mind, MouseFlow provides a solution that enables efficient and intuitive processing of dMRI datasets. This pipeline guarantees the reproducibility of local diffusion tensor measurements (DTI) and fODFs, as well as some tractography results, all without the need for complex installation steps. One of the central objectives of this pipeline is to promote high-performance and reproducible diffusion tractography processing, in line with the principles of open science. By adopting recognised industry standards, MouseFlow is helping to standardise practices and enhance the quality of research in this field. MouseFlow is the first turnkey solution dedicated to standardising from diffusion MRI data processing to diffusion metrics and tractography for the preclinical imaging community. Initially developed for ex vivo mouse brain analysis, MouseFlow will be soon extended to support in vivo applications. By providing a universal and reproducible dMRI pipeline, MouseFlow aims to enable longitudinal studies and foster the widespread adoption of standardised practices in rodent brain connectivity research.
Elise COSENZA (Bordeaux), Arnaud BORÉ, Alex VALCOURT CARON, Valéry OZENNE, Aurélien TROTIER, Maxime DESCOTEAUX, Sylvain MIRAUX, Laurent PETIT
14:02 - 14:04
#47778 - PG152 Studying cortical lamination in the developing brain with quantitative MRI.
PG152 Studying cortical lamination in the developing brain with quantitative MRI.
Throughout development, the human brain's cortex undergoes profound changes that drive the emergence of various cognitive and sensorimotor skills. Structurally, the cortex organizes into six cytoarchitectonic layers with distinct feedforward and feedback connections. The differentiation of cortical layers is reflected by varying levels of myelin and iron, supporting processes like neural communication, energy production and neurotransmitter regulation[1,2]. The myeloarchitecture and iron distribution of cortical layers vary across brain regions to support specialized functions, making the study of cortical lamination during development crucial for understanding brain structure function interaction[3].
In the newborn brain, myelin and iron levels are low, but they increase dramatically during the lifespan. Flechsig, the originator of the view that myelination correlates with functional maturation, hypothesized that regions more myelinated at birth develop faster postnatally[4]. Iron’s role in shaping functional development was studied by Hallgren and Sourander, who observed a rapid increase in iron in the first two decades, followed by slower growth[5]. However, these findings, based on cross-sectional qualitative assessments of selected regions, were never tested quantitatively and longitudinally across cortical layers and the whole brain.
Quantitative MRI (qMRI) revolutionized non-invasive brain imaging, offering biophysical parametric measurements of brain microstructure. Multiparametric mapping (MPM) combines qMRI parameters for whole-brain exploration of myelin, iron and water content, with submillimeter resolution[6]. Cortical lamination has been characterized with qMRI in adults, by sampling MPM measurements from deep white matter to the pial surface, creating cortical profiles[3,7–10]. However, this multiparametric approach has not yet been implemented in infant brains and compared to adults. Thus, the rates of myelination and iron accumulation in infancy and their relation to adult values across cortical depths and functional systems remain largely unexplored.
Here, we present an acquisition protocol and data analysis pipeline for laminar MPM in the infant cortex. By comparing infant and adult values we explore layer-dependent microstructural development. In addition, we show the generalizability of our approach by comparing different quantitative parametric methods.
Healthy infants (N=15, 9-15 months old) were scanned on a Siemens 3T Prisma MRI scanner during natural sleep (1 mm isotropic resolution). Adult participants (N=10) were scanned on a Siemens 3T Connectome scanner (0.8 mm isotropic resolution).
MPM protocol[11]: 3D multi-echo gradient-echo scans with T1 (FA=21°), PD (FA=6°), and MT (FA=6°) weightings, TR=24 ms, 8 equidistant echoes (TE=2.4–18.4 ms), reduced to 5 for MT-weighting in infants and 6 in adults, BW=488 Hz/Px, FOV=256 mm, 2x2 CAIPI (infants)/ GRAPPA (adults). Spin-echo and stimulated echo (SESTE) images for B1+ mapping [12].
Magnetization-prepared two rapid-acquisition gradient-echo (MP2RAGE): FA=4° & 5°, TI=700 & 2500 ms[13].
Relaxation rates (R1, R2*), magnetization transfer saturation (MTsat), and B1+ maps were calculated using the hMRI toolbox[11]. The cortical surfaces were reconstructed using FreeSurfer based on MP2RAGE in infants (MPRAGE for adults) and registered to MPM. We generated R1, R2*, and MTsat maps for infants and adults. Figure 1 illustrates the obtained qMRI maps. Figure 2 shows a comparison of qMRI measurements for the whole brain, revealing increases in R1, MTsat, and R2* values with age (32%, 140% & 47% correspondingly in the white-matter). Next, by comparing the cortical profiles in the occipital cortex of two representative subjects, we found parameter-specific changes, suggesting distinct maturation patterns for different microstructural properties. Finally, we compared R1 estimations from MPM and MP2RAGE in the same infant, finding strong agreement, with slightly higher R1 values from MP2RAGE (Fig. 4a). Cortical profiles were similar across R1 methods in infant but distinct from the adult profile (Fig. 4b). This study demonstrates the feasibility of using qMRI-based MPM to examine cortical microstructural development in infants. While previous studies mapped adult cortical profiles, our work extends this multiparametric analysis to infants, capturing early myelination and iron accumulation across cortical depths for the whole brain. The agreement between MPM and MP2RAGE-derived R1 values supports the robustness of this approach for developmental research. Expanding this framework to larger cohorts could elucidate microstructural development across functional networks. We presented a qMRI framework for investigating cortical lamination across development, which may greatly enhance our understanding of lifespan changes in myelination, iron accumulation, and their functional relevance.
Shir FILO (Leipzig, Germany), Cheslie KLEIN, Juliane DAMM, Luke EDWARDS, Kerrin PINE, Angela FRIEDERICI, Nikolaus WEISKOPF, Charlotte GROSSE WIESMANN, Evgeniya KIRILINA
14:04 - 14:06
#46883 - PG153 Progress in direct mapping of myelin T1 in ex vivo brain with and without deuteration.
PG153 Progress in direct mapping of myelin T1 in ex vivo brain with and without deuteration.
T1 is an established parameter in quantitative MRI due to its excellent contrast between white matter (WM) and grey matter (GM) and its sensitivity to pathologies [1]. Estimates of the aqueous T1 in WM at 3T span a wide range of 700-1735ms across sequences and signal models [2]. This is attributed to magnetization transfer (MT) between the water and macromolecular (MM) proton pools [3-6]. MT effects depend on the inherent T1 times of the pools as well as their magnetization following RF pulsing and thus sequence parameters [7,8]. To obtain direct access to myelin T1 and probe how MT affects T1 in different pools, variable flip angle (VFA) short-T2 MRI was used to directly measure the T1 of myelin and non-myelin MM as well as water components in a deuterated brain sample [9]. This preliminary study was limited by B1 inhomogeneity and relatively small FA range. Moreover, ex vivo measurements suffer from sensitivity to tissue degeneration and external parameters such as temperature. Here, we present methodological improvements to this approach, including the extension of the FA range, B1 mapping and signal stability monitoring. These are applied to both deuterated and untreated porcine brain.
Two slices of porcine brain were stored frozen and thawed before scanning. One sample underwent an almost complete exchange of H2O with D2O to minimize water signal. A second sample was left untreated. Single-point imaging (SPI) was performed on a 3T Philips Achieva system with dedicated short-T2 hardware, including a high-performance gradient (220mT/m, 100% duty cycle) [10]. SPI was acquired at 14 different TE=33-2067us for sets of FAs ranging from 2 to just above 20 deg. Flip angles maps were estimated via scaling of high-resolution reference B1-efficiency maps acquired in doped water with a global B1-efficiency measured in the tissue sample. Multi-TE series at FA ~9 were repeated throughout data acquisition to monitor signal stability. The components were separated by voxel-wise fits of a 3-component model to each multi-TE series, resulting in three amplitude maps for each FA. They correspond to myelin (AU, ultra-short T2~5us) and non-myelin (AS, short T2~100us) MM and water (AW, long T2~50ms) [11-13]. The steady-state signal equation was fitted to amplitudes as a function of FA to estimate T1 for each component. Fig. 1 shows the signal component amplitude maps and the relative component amplitudes averaged over the green WM ROI. Fig. 2 shows T1 maps and averages over the WM ROI in Fig. 1. T1 in MM pools is shorter than water T1. Water T1 is at the low end of the range of previous estimates. T1 times are longer in D2O. In Fig. 3 signal amplitude averages over the ROI from Fig. 1 are plotted as a function of FA with curves corresponding to best fits of the steady-state equation. Solid circles represent data included in the fit, points shown as open circles were used to monitor signal stability. The water component in D2O and the myelin component in H2O show systematic deviations from the signal equation. Open circle data from Fig. 3 are plotted as a function of scan time in Fig. 4. Changes in the MM pool amplitudes over time are small compared to changes across FA. In D2O, water signal increases over time, while changes in H2O are negligible compared to changes over FA. In D2O the effect of MT on MM pools is minimized and T1 estimates are expected to correspond to intrinsic T1. The obtained values are in good agreement with previous estimates from multi-pool modeling [5,7,14-16] and UTE imaging [17,18] and significantly shorter than T1=1s widely used in quantitative MT [19]. In H2O, MT affects T1 relaxation in the MM components, resulting in a longer apparent T1 and a deviation from the steady-state equation. A deviation is also observed for the water pool in D2O, where MT effects are expected to be enhanced by the increased relative size of the MM pool. This also explains the decrease in the apparent T1 compared to the H2O sample which is consistent with previous findings [20]. The water T1 might be further reduced due to the presence of significant myelin water signal at short TE, which has been reported to have shorter T1 than intra/extracellular water [16]. Signal variation over time is small compared to changes across FAs and is thus not expected to bias T1 estimates except for the water pool in D2O. Direct T1 mapping of the water as well as myelin and non-myelin MM pool has been successfully performed in a deuterated and an untreated brain sample. We improved upon our previous results of the intrinsic T1 of the myelin and non-myelin MM, obtaining T1^U=226±7 and T1^S=236±19. Observed deviations from the steady-state signal equations in the smaller pools as well as the increase in apparent T1 following deuteration can be attributed to the presence of MT effects that are modulated by pool sizes.
Lara Maria BARTELS (Zurich, Switzerland), Emily Louise BAADSVIK, Markus WEIGER, Benjamin Victor INEICHEN, Klaas Paul PRUESSMANN
14:06 - 14:08
#45942 - PG154 A temperature correction model for MRI parameters in deep gray matter substructures using real-time forehead temperature.
PG154 A temperature correction model for MRI parameters in deep gray matter substructures using real-time forehead temperature.
A major limitation of postmortem (PM) in-situ MRI is the variability in brain temperature at the time of image acquisition as it affects multiple MRI parameters [1]. To ensure reliable group comparisons, temperature-related alterations in MRI parameters must be corrected. A previous study developed a temperature correction model based on real-time forehead temperature measurements during the MRI acquisition [2]. This model can be applied to correct MRI parameters in white matter, cerebral cortex, and deep gray matter (DGM) as a whole. Nevertheless, for the investigation of neurodegenerative diseases, the evaluation of specific DGM substructures, such as hippocampus or brainstem is paramount [3,4].
The aim of this study was to develop a temperature correction model for MRI parameters in DGM substructures using non-invasive real-time forehead temperature measurements.
All procedures were approved by the local ethics committee. PM in-situ (brain not extracted from the skull) whole-brain MRI scans were conducted in 17 forensic cases (age at death = 56.1 ± 14.8 years; four females, 13 males), all with an autopsy request by the local prosecutor. Prior to the scan, five of the subjects were stored at room temperature (19 °C), while twelve were stored in a cooling chamber at 4 °C.
During the MRI scan, the forehead temperature was continuously assessed every 10 s using an MR safe surface temperature probe with a measurement accuracy of ±0.2 °C (custom build product, Testo AG, Mönchaltorf, Switzerland). For further analysis, the temperature was averaged for each sequence.
The scans were performed at 3T (Siemens MAGNETOM Prisma), including the following sequences: Diffusion-weighted single-shot echo-planar imaging DTI sequence to quantify MD and FA; multi-contrast spin echo sequence with 12 different TEs to quantify T2; multi-echo gradient echo sequence with 12 different TEs to quantify T2*; inversion recovery spin echo sequence with 6 different inversion times to create the DGM masks and to quantify T1.
FSL FIRST [5] was used to create masks for the DGM substructures caudate, putamen, pallidum, hippocampus, amygdala, thalamus, and brainstem. After correcting for eddy currents, FSL’s dtifit command was used to generate FA and MD maps. T2 and T2* maps were generated using a voxel-wise two-parameter mono-exponential single decay fit, while T1 was calculated voxel-wise using a biexponential fit with three parameters (M0, p, T1) and T2 of the corresponding voxel using python. The factor p accounts for the B1 error leading to a non-ideal spin inversion.
All statistical analyses were performed using python. The MRI parameters were averaged for each tissue type. The temperature dependence of each MRI parameter was assessed by fitting a linear model to the data, as proposed by Nelson et al. [6]. Further, 95 % confidence intervals of the linear fits were determined. A Pearson’s p-value ≤ 0.05 was considered statistically significant. The Pearson correlation coefficient (r) was used to assess the strength and direction of the linear relationship between the MRI parameter and temperature. Fitting a linear model to the MRI data revealed statistically significant temperature dependencies for MD across all investigated DGM substructures except for pallidum, and for T1 in the putamen, pallidum, hippocampus, and brainstem. Among these cases, the correlation was positive, except for T1 in the putamen, where a negative correlation with temperature was observed. No significant effect of temperature was observed on FA, T2, and T2*. For the statistically significant cases, the fitted linear models and the corresponding statistical parameters are shown in Table 1. The linear models fitted to the data, with MRI parameters plotted against the mean forehead temperature per sequence, are shown in Figure 1 (FA and MD), Figure 2 (T1), and Figure 3 (T2 and T2*). The objective of this study was to develop a temperature correction model for MRI parameters in DGM substructures using real-time forehead temperature. In PM in-situ studies, it is essential to apply temperature correction to the MRI parameters that showed a significant linear correlation with temperature. Such temperature corrections can be performed using the intersect a and slope b derived from the fitted linear models. However, this approach is limited by the assumption of a uniform temperature distribution within the brain. Nevertheless, forehead temperature can be measured non-invasively and in real-time, without delaying MRI scans or introducing additional temperature-related effects. This method is therefore well-suited for practical use. This study demonstrated a linear correlation of MD and T1 with temperature in several DGM substructures. The fitted linear models can be used to correct PM MRI parameters for temperature, which is a crucial prerequisite for performing group comparisons, for example in the context of biomarker development for neurodegenerative diseases.
Dominique NEUHAUS, Dominique NEUHAUS (Basel, Switzerland), Eva SCHEURER, Claudia LENZ
14:08 - 14:10
#46103 - PG155 Microstructure differences in metastasis, glioma and healthy brains using neurite orientation dispersion and density imaging.
PG155 Microstructure differences in metastasis, glioma and healthy brains using neurite orientation dispersion and density imaging.
Brain tumors can cause not only localized tissue disruption but also widespread alterations in brain structure and function [1]. While conventional MRI focuses on identifying tumor mass and peritumoral edema, it may miss subtle microstructural changes that can impact clinical outcomes. Neurite orientation dispersion and density imaging (NODDI) enables a more detailed assessment of brain tissue by quantifying orientation dispersion (ODI), neurite density (NDI) and free water fraction (FWF) [2]. Prior studies focused on tumor and peritumoral areas with limited investigation of brain-wide effects. Therefore, this study aimed to systematically compare microstructural alterations in both hemispheres of metastasis and glioma patients with those of healthy controls. This helps to understand how tumors and edema affect brain microstructure beyond the visible lesion.
We included four patients with morphologically atypical brain metastases, three patients with gliomas and nine healthy controls (Table 1). All tumors were restricted to one hemisphere. Brain MRI scans were performed at a 3 T (MAGNETOM Skyra, Siemens Healthineers, Erlangen, Germany) scanner using a 64-channel head coil. The scan protocol consisted of a diffusion weighted multi-shell echo-planar imaging diffusion tensor imaging (DTI) sequence (b = 1000 and 2000 s/mm², 30 diffusion directions, 5 b = 0 s/mm², TE = 97 ms, TR = 6700 ms, isotropic resolution of 2 mm³), a reverse phase-encoded DTI sequence (b = 0 s/mm²) and a magnetization prepared rapid gradient echo sequence (MPRAGE; TE = 2.12 ms, TR = 1690 ms, flip angle = 8°, isotropic resolution of 0.8 mm³). Diffusion data were preprocessed using TractoFlow [3]. To compute the maps of ODI, NDI and FWF, the Watson model in the cuda diffusion modelling toolbox (cuDIMOT; [4]) was used. In 3D Slicer [5,6], ball-shaped regions of interests (diameter approx. 1.5 cm) were manually placed in normal appearing WM of the affected hemisphere. Freesurfer [7] was applied to segment the contralateral brain hemisphere of the patients and the whole brain of the controls into white matter (WM), deep gray matter (DGM) and cortex. Analyses of covariance (ANCOVA; age as covariate) were performed using MATLAB [8]. Boxplots illustrating the distribution of ODI, NDI and FWF values for normal appearing WM in the affected hemisphere, contralateral hemisphere and controls are presented in Figure 1. Table 2 summarizes the ANCOVA results.
In WM of the affected hemisphere, both metastasis and glioma patients showed statistically significantly higher FWF compared to controls. Additionally, glioma patients exhibited increased NDI relative to controls. In the contralateral hemisphere, FWF was significantly higher in metastasis patients in WM and cortex and in glioma patients in WM and DGM compared to controls. ODI was significantly lower in the contralateral cortex of metastasis patients and in contralateral DGM of glioma patients.
Age was a significant confounding factor for FWF in the contralateral cortex of metastasis patients as well as for FWF in the contralateral DGM and cortex and ODI in the contralateral WM and cortex of glioma patients.
Significant interactions of group and age were observed for NDI in the contralateral cortex and FWF in the contralateral WM and cortex of metastasis patients as well as for FWF in the contralateral DGM and ODI in the contralateral WM of glioma patients. Increased FWF in the affected hemisphere compared to controls can be attributed to the presence of edema or extracellular water accumulation, even in normal appearing WM [9,10]. The space occupying nature of gliomas may compress surrounding tissue, resulting in elevated NDI in the affected hemisphere [11]. Also in the contralateral hemisphere, FWF is higher than in controls, suggesting fluid accumulation not only in the affected but also in the contralateral hemisphere [12]. Decreased ODI in cortex and DGM may reflect altered neuronal connectivity or compensatory reorganization, reducing dendritic complexity.
Age emerged as a significant factor, explaining a considerable portion of the variability in both ODI and FWF in the contralateral hemisphere beyond group effects alone. Moreover, the significant group-by-age interactions highlight the non-uniform influence of age on NODDI parameters across patients and controls. This finding underscores the importance of accounting for age when interpreting microstructural metrics. Our findings highlight the sensitivity of the NODDI model to subtle, widespread changes in brain microstructure. Edema and microstructural alterations are not confined to the tumor vicinity, emphasizing the importance of comprehensive brain assessment in treatment planning and disease monitoring. Larger studies are needed to strengthen these preliminary findings, clarify NODDI parameter differences between low- and high-grade gliomas and explore gender-specific differences.
Melanie BAUER (Innsbruck, Austria), Stephanie MANGESIUS, Michaela WAGNER, Johannes KERSCHBAUMER, Daniel PINGGERA, Astrid GRAMS, Elke R. GIZEWSKI, Christoph BIRKL
14:10 - 14:12
#47823 - PG156 Iron load and decreased myelination of deep grey matter nuclei in leukodystrophies, new insights using DECOMPOSE-QSM.
PG156 Iron load and decreased myelination of deep grey matter nuclei in leukodystrophies, new insights using DECOMPOSE-QSM.
Leukodystrophies (LD) are a group of rare, genetic, progressive disorders characterized by the occurrence of white matter (WM) changes in the brain. Due to the highly heterogeneous spectrum and rarity of LD, there is still a lack of MRI quantitative biomarkers useful to guide diagnosis, monitor progression, and innovative therapeutic approaches. Although WM degeneration is the hallmark of LD, the involvement of deep gray matter (DGM) structures remains largely unexplored.
In other neurodegenerative disorders[1,2,3], Quantitative Susceptibility Mapping (QSM) has been proved to be useful as a quantitative biomarker and recently some algorithms for the separation of paramagnetic and diamagnetic susceptibility sources, such as DECOMPOSE-QSM[4] and ????-separation[5], allow to distinguish the contribution of iron and myelin colocalized within the same voxel. This pilot study aims to use DECOMPOSE-QSM to explore the patterns of myelination and iron deposition in DGM nuclei in leukodystrophies.
Five patients with LD (23-30yo, 4M, carrying mutations in the genes PLP1, ERCC2, GFAP, POLR3, MORC2) and nine healthy controls (HC) (30-40yo, 5M) underwent an MR exam on a 3T GE Signa Premier scanner. The protocol included a multi-echo GRE with whole-brain coverage, a T2-weighted FLAIR for lesion detection, and a T1-weighted MPRAGE for anatomical reference. GRE magnitude images were skull-stripped using FSL-bet[6] and AFNI-3dAutomask[7]. Susceptibility maps were computed from the GRE phase via STISuite[8] using laplacian phase unwrapping[9], V-SHARP[10] background field removal, and iLSQR[11,12] for dipole inversion. Paramagnetic and diamagnetic components of susceptibility (PCS and DCS) were estimated using DECOMPOSE-QSM4.
By using ANTs, the GRE magnitude image and the T1-weighted image of HCs were coregistered and used to create a study-specific template, then warped to the CIT168 template[13]. From this atlas, we derived the ROIs for caudate nucleus (Ca), putamen (Pu), globus pallidus (GP), red nucleus (RN), and substantia nigra (SN) by setting a 0.4 probability threshold to the probabilistic labels and eroding by one voxel. In one exception, the dentate nucleus (DN) was manually drawn on the study-specific template.
Dysmorphism and atrophy hindered accurate registration of patients to the template. Hence, all ROIs were manually segmented on the T2*-weighted image of each patient.
Average QSM, PCS, and DCS were computed for each ROI and each subject and were age-corrected using linear regression when a significant Pearson’s correlation with age was found (p<0.05). Values of left and right ROIs were averaged. These values were compared across groups using the Mann-Whitney U test (significance threshold p<0.05 after False Discovery Rate correction). To explore the susceptibility pattern of single patients, they were individually compared to the control population by computing the t-score for each ROI and each map. Figure 1 displays the anatomical FLAIR images of all patients compared to one control subject. Figure 2 shows QSM, PCS, and DCS maps for a GFAP patient and an age-matched control. Age effect was reported in several nuclei for QSM and PCS, but not for DCS. In Figure 3A, we report an increase in QSM in LD patients compared to HC in GP and Pu, while in Figure 3B, in comparison, PCS was increased not only in these ROIs but also in SN, potentially indicating higher PCS sensitivity to iron overload with respect to QSM. In Figure 3C, we highlight the lower absolute DCS in GP, Pu, and DN, suggesting a reduction of myelination. Figure 4 shows the t-score analysis of individual subjects revealing different patterns of iron load and myelination in different pathologies and individuals, showing different degrees of degeneration of basal ganglia, midbrain, and cerebellum. DGM nuclei involvement may be present in individuals with leukodystrophies. In this pilot study, we observed susceptibility alterations due to increased iron load concurrently with decreased myelination in DGM. While alterations of the WM are well described in these patients, myelin reduction in DGM nuclei remains underinvestigated and iron deposition has only been reported anecdotally in animal models[14] and in other neurodegenerative disorders[15]. It is of note that each patient presented a unique distribution of these alterations. This individual specificity in the pattern distribution should be investigated to underline possible genotype-imaging phenotype correlations. QSM mapping may represent an important and clinically applicable tool for the quantitative assessment of leukodystrophies in DGM. The investigation of these patterns of iron and myelin spatial distributions paves the way for new diagnostic hints and specific follow up biomarkers.
Marta LANCIONE, Matteo CENCINI, Bianca BUCHIGNANI, Rosa PASQUARIELLO, Domenico MONTANARO, Chunlei LIU, Roberta BATTINI, Laura BIAGI, Michela TOSETTI (Pisa, Italy)
14:12 - 14:14
#47655 - PG157 Intracranial volume loss and skull thickening in metachromatic leukodystrophy and multiple sclerosis.
PG157 Intracranial volume loss and skull thickening in metachromatic leukodystrophy and multiple sclerosis.
Intracranial volume (ICV) is often used as a normalization factor in volumetrics and is considered stable in adults [1]. We noticed thick skulls on MRI scans of leukodystrophy patients. These are rare, genetic disorders, affecting mainly the white matter, sometimes leading to atrophy at a young age. This observation prompted us to quantify the skull thickness in patients with metachromatic leukodystrophy (MLD), a leukodystrophy characterized by early-onset atrophy [2]. Additionally, we investigated MRI scans of people with multiple sclerosis (pwMS), to investigate whether similar skull changes could occur in a non-genetic disease. In this study, we aimed to quantify skull thickness and ICV in patients with MLD and in pwMS, and the potential relation between skull thickness and ICV.
We retrospectively analyzed cross-sectional and longitudinal MRI scans of participants with MLD (n=32, 11 male, scans=136, median age first scan=14.1 [IQR 7.9-25.7] years), MS (n=232, 78 male, median age first scan=47.3 [IQR 39.6-55.4] years, scans=431), and controls (n=140, 68 male, median age first scan=31.2 [IQR 10.7-48.9] years, scans=319). A flow-chart of the analysis is shown in Fig. 1. ICV was estimated from 3D T1-weighted images using SynthSeg [3]. To determine skull thickness skull surfaces were extracted from 3D T1-weighted images, using FSL BET’s betsurf function, supplemented with T2-weighted images when available [4]. Skull bases were removed, and point clouds (5×10⁵ points) were generated with BrainCalculator [5]. Skull thickness was calculated as the median Euclidean distance between points between the inner and outer skull surfaces. The MLD and control groups were split into participants below and above 20 years to account for natural ICV and skull growth. In the pwMS group, all participants were older than 20 years. For both age groups separately, linear regression models were fitted to calculate slopes (Δ) in ICV and skull thickness per year for each subject. Scatterplots of ICV and skull thickness as a function of age are shown in Fig. 2A and B, respectively. Slopes in ICV and skull thickness are shown in Fig. 3. Both ICV and skull thickness showed natural growth in young (<20y) controls. In young MLD participants, ICV decreased (-18.8 ± 22.4 mL/year, p<0.001). Above age 20y, ICV and skull thickness remained stable in controls. In comparison to controls, we observed ICV loss in MLD participants (-4.01 ± 8.29 mL/year, p=0.004) and in pwMS (-2.99 ± 2.69 mL/year, p<0.001), as well as skull thickening (MLD: 0.16 ± 0.14 mm/year, p<0.001, pwMS: 0.04 ± 0.09 mm/year, p=0.013). The relation between ICV and skull thickness in adult groups is shown in Fig. 2C. Negative correlations were found between ICV and skull thickness in both disease groups (MLD: -19.39mL/mm, p<0.001, pwMS: -13.16 mL/mm, p<0.001), but not in controls (p=0.222). These findings challenge the assumption of a stable ICV after reaching adulthood and suggest an adaptive skull response to atrophy. In (cross-sectional) analyses using normalized volumes, ICV shrinkage may potentially underestimate atrophy. Further research is needed to explore the underlying mechanisms of ICV shrinkage and skull thickening, and its potential impact on volumetric outcomes to make more accurate evaluations of brain atrophy progression. This study provides evidence that in MLD and pwMS skull thickening and ICV shrinkage occur, challenging the assumption of a stable ICV in adults, and even before reaching adulthood.
Guus VORST, Petra POUWELS (Amsterdam, The Netherlands), Nicole WOLF, David VAN NEDERPELT, Frederik BARKHOF, Marjo VAN DER KNAAP, Menno SCHOONHEIM, Eva STRIJBIS
14:14 - 14:16
#47678 - PG158 Investigating the relationship between sickle cell anaemia and brain tissue conductivity in Tanzanian children using MR-EPT at 1.5T.
PG158 Investigating the relationship between sickle cell anaemia and brain tissue conductivity in Tanzanian children using MR-EPT at 1.5T.
Sickle cell anaemia (SCA) is a genetic blood disorder causing haemoglobin to polymerize and red blood cells to adopt an abnormal sickle-shape. SCA is known to impact normal neurocognition, and poses serious risks such as haemorrhagic or ischaemic stroke [1]. The relationship between SCA and magnetic susceptibility has recently been studied [2], but its effect on tissue electrical conductivity (σ) remains unknown.
σ is an intrinsic tissue property determined by the concentration (and mobility) of ions [3]. This can be non-invasively measured via phase-based Electrical Properties Tomography (EPT), which derives σ from the MRI transceive phase (φ0), using the integral form of the truncated Helmholtz equation [4]. Here, we used a previously optimised EPT pipeline [5,6] to test whether SCA has an impact on brain tissue conductivity. In children with SCA, we also investigated whether the presence of silent cerebral infarcts (SCIs) or vasculopathy affects conductivity [7].
Using an optimised EPT pipeline [5,6], conductivity maps were generated for a cohort of 231 Tanzanian children: 181 with SCA and 50 healthy controls (HCs), aged 13.1 ± 4.0 and 10.9 ± 3.4 years, respectively, 109/122 male/female.
MRI acquisition: T2*-weighted multi-echo 3D GRE and T1-weighted MPRAGE were acquired at Muhimbili National Hospital, Tanzania, on a 1.5T Phillips Achieva system using either an 8-channel or birdcage RF coil. 3D-GRE had: 5 echoes, TE1 = 4.28 ms, ΔTE = 4.94 ms, TR = 27.4 ms, resolution = 1.458 x 1.458 x 1.5 mm3, bandwidth = 287 Hz/pixel, FA = 15˚.
Analysis: Grey matter (GM), white matter (WM) and cerebrospinal fluid (CSF) were segmented from the T1-weighted image using FSL-FAST [8]. 15 smaller regions of interest (ROIs) were segmented from the same T1-weighted image using SynthSeg [9]. To account for age differences between SCA and control groups, age correction was applied to σ values by regressing out linear age effects. For regional analysis, the median conductivity value was calculated, excluding negative values and values > 10 S/m, as these were considered physically implausible and erroneous. Groupwise comparisons (two-sample t-tests) were performed between age-corrected median σ in SCA vs HCs. The impact of sex and age on σ was investigated using ANOVA. In SCA, the effect of SCIs and vasculopathy on σ was assessed using two-sample t-tests. Fig. 1 shows a representative conductivity map and corresponding ROI segmentation.
Fig. 2 shows groupwise comparisons of median conductivity, across tissue types. Table 1 shows the median conductivity values in all tissue types and ROIs, averaged across all subjects, alongside t-test p-values without and with age-correction. With age-correction, no significant differences were observed between SCA/HCs, in any tissue type or ROI.
Fig. 3 illustrates a significant negative correlation between σ and age in WM, in the SCA group only (not HCs). Specifically, this age-related effect was significant in the cerebral WM (p=0.002) but not in the cerebellar WM. No significant conductivity-age correlations were found in CSF, GM, or other ROIs.
No significant association was observed between σ and the presence of SCIs or vasculopathy. This work represents the first application of MR-EPT to study SCA, offering novel insights into a complex and relatively understudied disease. Our findings indicate that the presence of SCA does not significantly affect brain tissue conductivity.
WM conductivity was found to significantly decrease with age in SCA, supporting the hypothesis that SCA acts as an accelerated aging syndrome [10]. The fact that this effect occurred only in WM may relate to the widespread WM integrity loss previously associated with SCA [11], which may worsen over time.
Future work will consider relationships between σ and other clinical variables, such as oxygen saturation. We used an optimised EPT pipeline to investigate the effect of SCA on brain tissue conductivity, in Tanzanian children at 1.5T. The presence of SCA had no significant impact on conductivity. However, in SCA, WM conductivity was negatively correlated with age. By optimising EPT at 1.5T, this work presents an exciting opportunity to study SCA and other diseases affecting the brain, particularly in less medically developed parts of the world.
Philippa SHA (London, United Kingdom), Jierong LUO, Oriana ARSENOV, Mitchel LEE, Mboka JACOB, Dawn SAUNDERS, Fenella KIRKHAM, Karin SHMUELI
14:16 - 14:18
#46428 - PG159 Regional brain volume alterations in FGFR-related craniostenosis: a normative modeling approach.
PG159 Regional brain volume alterations in FGFR-related craniostenosis: a normative modeling approach.
Craniostenosis is a cranial malformation caused by the premature closure of some sutures [1], and may be of syndromic origin, as in the rare Crouzon syndrome (CS, 1/50,000 births), Muenke syndrome (MS, 1/30,000 births) and Apert syndrome (AS, 1/65,000 births) [2,3]. These are caused by mutations in the FGFR-2 and FGFR-3 genes [3], which play critical roles in skull and brain development, and have been associated with variable neurodevelopmental outcomes. In this study, we investigated regional brain volume growth and scaling in these FGFR-related syndromes using a normative modeling approach.
We analyzed 65 patients, including 23 pre-operative CS, 7 post-operative (1st step) CS, 4 pre-operative AS, 6 post-operative (1st step) MS and 23 post-operative non-syndromic (NS) patients, imaged at Hôpital Necker-Enfants Malades (mean age=5.46 years, M/F=38/25). A control group of 130 typically developing children (from Hôpital Necker, the Baby Connectome Project, and Neurospin) was included (mean age=5.99 years, M/F=72/58), with 25% of site-matched controls (Hôpital Necker).
3D isotropic T1-weighted MRI scans were segmented into 6 hemispheric and 3 subcortical structures using AssemblyNet [4] and Morphologist [5] pipelines. Regional volumes were extracted. Lateralization indices of volumes were calculated and did not show significant differences so volumes of homologous left and right regions were summed. ComBat harmonization was applied to correct for site effects then for sex.
First, transversal growth curves were estimated in controls using a Turner's model [6] for each volume of interest. Homoscedasticity of residuals was confirmed (using Breusch-Pagan and 5-slot Levene's tests) which allowed for reliable z-score estimation. Patient data were evaluated within this normative framework to assess deviations from typical development. We used a Shuffle and Split procedure (a permutation-based test able to accommodate small sample size) to assess mean deviations at the group level. Additionally, the proportion of abnormal individuals (below the 10th or above the 90th percentile) was estimated and tested against the proportion observed in controls using a proportion test.
Second, age independent volumes were obtained by projecting all subjects to 18 years of age, assuming preserved z-scores. Static scaling curves (with respect to brain size, i.e., total brain volume) were then estimated using a power-law model. Homoscedasticity of residuals was confirmed. Normative scaling analysis was performed using the same permutation and proportion tests as previously described for growth. In pre- and post-operative patients with CS, regional brain volumes were expected for age (growth) and brain size (scaling), except for enlarged lateral ventricles (corrected p<10⁻⁹). In patients with AS, total brain volume, cerebellum, ventricles, temporal, parietal, and central volumes were increased for age (corrected p=10⁻3, 10⁻7, 10⁻7, 10⁻7, 0.02, 10⁻7 respectively), but notably not frontal volume. With respect to brain size, temporal and cerebellar volumes were oversized (corrected p<10⁻7 for both) while frontal volume was undersized (meaning under-scaled). In patients with MS, temporal volume was increased for age (corrected p=10⁻6) and oversized with respect to brain size (corrected p=10⁻7), while frontal volume was under-scaled (corrected p=10⁻7). Postoperative NS patients showed brain volumes consistent with controls for age but a slight frontal oversizing and temporal undersizing relative to brain size (corrected p=0.02 and 0.03 respectively). In CS, aside from ventricular enlargement, brain volumes showed no deviation from typically developing individuals. Given the high clinical and genetic heterogeneity of CS, with variable suture closure and diverse FGFR2 mutations, some subgroups might exhibit more pronounced neuroanatomical deviations not detected due to limited statistical power. By contrast, patients with AS and, to a lesser extent, patients with MS showed brain volumetric anomalies. Interestingly, the disproportions observed in the AS macrocephalic brain (undersized frontal and oversized temporal volumes for brain size) are also observed in the normocephalic MS brain. This finding may have pathophysiological significance considering that these syndromes both have high genetic homogeneity but in two different FGFR genes. These neuroanatomical patterns suggest that the mutation itself, beyond cranial morphology, plays a key role in brain development. Our results provide new insights into the neuroanatomical consequences of FGFR-related craniosynostoses, revealing that both differential brain growth and scaling can be altered in genetically homogeneous (MS and AS) forms and promoting further analyses to identify more subtle subgroup-specific alterations in heterogeneous (CS) forms. They also show that sensitive computational normative neuroanatomy can be achieved in such rare diseases using care-related MRI datasets.
Ombline DELASSUS (Paris), Lucas CHOLLET, Barbara YOUNGUI, Jérémy SADOINE, Giovanna PATERNOSTER, Jean-François MANGIN, Roman Hossein KHONSARI, David GERMANAUD
14:18 - 14:20
#47830 - PG160 Abnormalities in brain anatomy are potential biomarkers of deep brain stimulation efficacy in children with dystonia.
PG160 Abnormalities in brain anatomy are potential biomarkers of deep brain stimulation efficacy in children with dystonia.
Deep brain stimulation of the globus pallidus internus (Gpi-DBS) is a highly effective surgical symptomatic treatment for various forms of pediatric-onset dystonia. However, clinical outcomes remain highly variable among dystonia forms. Several factors may contribute to this variability, including structural brain differences, particularly in regions associated with the dystonia connectome. These changes can be studied through the analysis of preoperative magnetic resonance images (MRI) in dystonia patients undergoing Gpi-DBS. The aim of this study was to characterize a cohort of dystonia patients treated with Gpi-DBS and a healthy subject (HS) group, by comparing their MRI-derived volumetric and cortical thickness measures. Additionally, it sought to explore the relationship between these measures and clinical outcomes by comparing data from responders and non-responders to GPi-DBS.
A cohort of 34 Gpi-DBS-treated dystonia subjects and 65 HS was analysed. Both groups included participants aged 7–20 years. Patients outside this range or with necrotic brain lesions were excluded. The dystonia group includes preoperative MRI from consecutive subjects who underwent DBS surgery at Vall d’Hebron Hospital between 2020 and 2024 with a minimum follow-up period of six months. Gpi-DBS response was assessed by comparing pre- and post-intervention scores on the Burke-Fahn-Marsden Dystonia Rating Scale. A postoperative improvement of ≥25% was used to define responders.
The MRIs of the HS group were obtained from the public OpenNeuro repository. The 3D T1-weighted MRI were segmented using Fastsurfer, providing measures of cortical thickness, as well as volumetric estimations of global metrics and subcortical structures. Cortical thicknesses were averaged across both hemispheres, resulting in 31 measures. Volumes from the left and right hemispheres were summed and normalized by the total estimated intracranial volume to yield 18 volumetric measures for comparison.
To harmonize values across different acquisition protocols and scanners, the parametric version of the ComBat tool was used. A linear model was applied to compare each morphological variable between patients and HS. ANCOVAs were conducted to assess differences between responders and non-responders, and between each of these groups and HS, using age as a covariate in both analyses. Post hoc comparisons were performed using estimated marginal means, and multiple comparisons were corrected using the Benjamini-Hochberg test. All statistical analyses were conducted in RStudio. The dystonia cohort included 4 different image-acquisition protocols: one with a 1.5T magnet and three in three different 3.0T magnets. The HS cohort also included 4 acquisition protocols, all using different 3.0T scanners. In the comparison between patients and HS, 18 out of 31 cortical regions showed a significant decrease in cortical thickness in the dystonia group (ranging from 2% to 11%) when compared to HS, while 3 regions showed a 2-3% increase in dystonia subjects. Regarding volumetric fractions, 15 out of the 18 measures were significantly reduced in dystonia patients, ranging from 5% to 19%. Ventricular volume was markedly increased—by approximately 35%—in dystonia patients compared to HS. Furthermore, the thalamus, hippocampus and total subcortical gray fractions were significantly reduced in non-responders to GPi-DBS compared to both HS and responders, with reductions ranging from 10% to 18% and 7% to 10%, respectively. Several cortical regions in Gpi-DBS patients showed significant cortical thinning when compared to HS, including the sensorimotor cortex, as well as the occipital, cingulate and frontal medial regions.
Volumetric analyses further revealed significant reductions in key brain structures, including the cerebellum, thalamus, brainstem, hippocampus, amygdala, and several basal ganglia regions such as the globus pallidus, putamen, and nucleus accumbens. Global volumetric measures were also decreased in dystonia patients, including total cerebral white matter and both cortical and subcortical grey matter fractions. Importantly, significant differences in subcortical gray matter volumes were observed between GPi-DBS responders and non-responders, suggesting a potential link between these structural characteristics and treatment efficacy. The integration of clinical MRI data with publicly available datasets, coupled with the use of ComBat for harmonization across acquisition protocols, presents a viable approach for studying brain morphology in pediatric dystonia. The presence of widespread structural differences—particularly in regions traditionally associated with dystonia, such as the basal ganglia and motor cortex, but also extending beyond—indicates a broader impact of the disorder on brain structure. More specifically, subcortical volumetric differences may hold promise as potential biomarkers for predicting GPi-DBS outcomes in children with dystonia.
Villa-María ANA, Lucero-Garofano ÁLVARO, Alberich MANEL, Marcé-Grau ANNA, Rovira ÀLEX, Vázquez ÉLIDA, Delgado IGNACIO, Deborah PARETO (Barcelona, Spain), Pérez-Dueñas BELÉN
14:20 - 14:22
#47774 - PG161 Subcortical nuclei in the Action-Observation Network investigated at Ultra-high Field.
PG161 Subcortical nuclei in the Action-Observation Network investigated at Ultra-high Field.
This study investigates the involvement of subcortical structures, particularly the basal ganglia and thalamus, in the Mirror Neuron System (MNS) using ultra-high-field 7T fMRI. Ten participants performed an Action-Observation (AO) and Action-Execution (EXE) paradigm while undergoing fMRI scanning. The study aimed to determine whether subcortical regions exhibit MNS-like activity during these tasks.
Ten participants underwent MRI acquisition on a SIGNA 7T MR system (GE Healthcare, WI, USA). BOLD responses were acquired during an Action-Observation (AO) paradigm and an Action-Execution (EXE) paradigm. A 2D EPI-GRE sequence with 1mm3 voxels was implemented (TR/TE=3s/29.2 ms, FA=78, 114 slices, FOV=21cm, matrix=210x210, ARC acceleration=3.00, HyperBand acceleration=3). Three fMRI runs were performed in AO condition, and two in EXE condition (4’21’’, 87 volumes+4 dummies, for each run). In the AO paradigm, participants watched videos of object manipulations (VIDEO) or images of objects (PICTURE). In the EXE paradigm, participants alternated between grasping a ball close to their right hand (GRASP) or opening and closing their hand (MOT). In AO and EXE paradigms, PICTURE and MOT conditions were control conditions. Instructions were presented on MR-compatible goggles. Conditions alternated in a block design with 15’’ blocks and interleaved by rest periods. The MR protocol included anatomical images (3D 0.6mm3 resolution MPRAGE TR/TI=1.6/1.1 s, FA=8, FOV=22cm, matrix=340x340). fMRI data were pre-processed in AFNI [5-6]. The pipeline included slice-timing and motion correction, brain extraction, correction of distortion by gradient acceleration, spatial alignment to MNI and 8mm smoothing. A General Linear Model (GLM) was used to generate statistical maps, using the SPM gamma variate basis function as hemodynamic response. Acquisition conditions (VIDEO or PICTURE in AO; GRASP or MOT in EXE) were considered as regressors of interest. Six motion parameters, white matter and cerebrospinal fluid maps were noise regressors. To detect voxels differentially activated in the two conditions (VIDEO>PICTURE; GRASP>MOT), we used a general linear test in AFNI. Single subject data were analysed by concatenating runs, group analysis by performing a multiple linear regression. Overlapping activations across subjects were drawn by summing binary masks of significant voxels. Activations at the group level are in Fig.1 and 2 for AO and EXE condition, respectively, showing positive and significant activations with p<0.001 after FDR correction. Both figures present in separate panels activations associated with each condition (VIDEO and PICTURE in Fig.1, GRASP and MOT in Fig.2) and their contrasts. The contrast maps (VIDEO>PICTURE and GRASP>MOT) show the MNS representation in the brain including cortical areas (frontal, parietal, occipital and temporal lobes), cerebellum, thalamic nuclei and basal ganglia (caudate nucleus and putamen). Contrast maps were combined through a logical AND to search common activations. Fig.3 shows voxels with positive contrast. Shared activations were in temporal, parietal and frontal cortices, cerebellum and thalamic nuclei bilaterally. In the basal ganglia, few voxels were active in both conditions. To assess the impact of inter-subject variability, we calculated maps of overlap from single-subjects. Fig.4 shows results for AO and EXE paradigms (panel A and B respectively), showing superposition of voxel with positive contrast across subjects (warmer colours=more overlap). Subcortical structures show active voxels clustered in the thalamus and basal ganglia with low overlap, indicating high inter-subject variability and at the same time the feasibility of observing subcortical activation at the single subject level (an example is reported in panel C). We leveraged on the superior SNR of 7T scanners to explore subcortical structures during observation and execution of complex actions. Our results at the group level confirm cortical, cerebellar and subcortical activations during execution and observation of manipulative actions. Activations were visible at the single subject level, with low spatial overlap in subcortical structures, indicating intrinsic inter-subject variability, leading to an underestimation of the activations in the group analysis. Region-of-Interest based analysis might be more suited to reveal activation in small structures as subcortical nuclei [7]. Results show activations in the cortex and cerebellum, as well as in the basal ganglia and thalamic nuclei, as reported from previous studies at lower field [7]. Single subject data suggest subcortical activations can be detected on individual participants, which might be important in rehabilitative treatments involving the MNS, like Action Observation Therapy (AOT) [8-9]. These results add to the possible involvement of subcortical nuclei in the MNS and highlight the advantage of Ultra-High Field in investigating non-cortical brain structures.
Pierfrancesco AMBROSI (Pisa, Italy), Marta LANCIONE, Paolo CECCHI, Antonino ERRANTE, Giuseppe CIULLO, Graziella DONATELLI, Giuseppina SGANDURRA, Leonardo FOGASSI, Michela TOSETTI, Laura BIAGI
14:22 - 14:24
#47252 - PG162 Normal-appearing white matter microstructural alterations in watershed regions of coronary artery disease patients and their link to cognition.
PG162 Normal-appearing white matter microstructural alterations in watershed regions of coronary artery disease patients and their link to cognition.
Coronary artery disease (CAD) is the most prevalent form of cardiovascular disease [1]. In addition to its well‐established effects on the heart [2], CAD is also associated with an increased incidence of cognitive decline [3]. White matter microstructural integrity is essential to maintain cognitive function, and diffusion MRI has shown links between WM microstructure and cognition in aging [4] and small vessel disease [5]. Importantly, WM microstructural alterations in normal appearing white matter (NAWM) may contribute cognitive dysfunction [6,7]. Within WM, watershed regions may be especially vulnerable to ischemic injury [8,9] since this is where terminal arterioles of adjacent vascular territories meet. These regions are also known to be prone to hypoperfusion [10] and WM hyperintensity (WMH) [11]. A large array of techniques can be used to probe WM microstructure, including R1 and magnetic susceptibility from Quantitative Susceptibility Mapping (QSM) which are both quantitative microstructure markers. They are both predominantly sensitive to myelin in WM, with additional contributions from iron [12], a marker of microglial activation and inflammation [13]. Here, we explored the microstructural integrity of NAWM in CAD patients compared to healthy controls by assessing QSM and R1 maps in watershed and non-watershed NAWM. We hypothesized that NAWM in the watershed areas is adversely affected in CAD patients and that these microstructural alterations are associated with poorer cognition.
The study includes 43 CAD patients (age = 68.2 ± 8.7 years, 8 females) and 36 healthy controls (HC) (age = 64.1 ± 7.8, 10 females). Each participant underwent a cognitive assessment from which composite scores for executive function and verbal episodic memory were calculated[14]. MRI data were acquired on a 3T Siemens Skyra. A B1 map and gradient-echo (GRE) sequence with variable flip angles were acquired for calculating B1 inhomogeneities-corrected R1 [15]. An MPRAGE, and axial T2-FLAIR images were acquired for tissue segmentation. A 3D multi-coil multi-echo GRE phase and magnitude data were acquired for QSM, with flow compensation on the first echo [16]. The R1 maps were calculated using the hMRI-toolbox (v0.3.0) in Statistical Parametric Mapping [17]. The phase data of multi-coil ME-GRE data was combined and unwrapped using ROMEO [18]. X maps were reconstructed using TGV-QSM [19]. NAWM and WMH, are segmented using the BISON classifier [20]. A cerebral arterial territory atlas was applied to extract watershed regions at the intersection between the anterior (ACA), middle (MCA) and posterior cerebral artery (PCA) regions [21]. Linear models were utilized in R to assess the group comparison and the relationship between cognitive scores and regional average X and R1. Partial correlation analysis was done to explore the correlation between X and R1. P-values were FDR-corrected (a = 0.05). Table 1 shows the demographic data of the participants. There was no significant difference in cognitive scores between groups. CAD was associated with higher X and lower R1 in all watershed regions (Figure 2), but not in non-watershed regions. Similarly, in all watershed regions, we identified a negative relationship between verbal episodic memory and X, while MoCA and executive function were associated with higher R1 (Figure 1). Finally, Figure 3 shows that there is a significant negative correlation between X and R1. Elevated X and lower R1 in CAD were confined to NAWM in watershed regions, consistent with watershed regions being especially vulnerable to vascular dysfunction [9]. R1 in WM is mainly driven by myelin content [12], thus this lower R1 is consistent with demyelination rather than iron deposition (which would elevate R1). Moreover, the significant negative correlation between R1 and X indicates that the main source of contrast is demyelination rather than iron deposition which may occur spontaneously in damaging NAWM [7]. If iron accumulation was dominant, we would expect a weak or even positive correlation. The association between executive function and MoCA scores with lower R1 is consistent with demyelination [22]. Poorer verbal episodic memory associates only with higher X, not R1. This indicates that there may be an additional component related to iron deposition in the relationship between microstructure and this cognitive domain, given the fact that myelination and iron deposition both contribute to higher X (Figure 3 bottom panel). This may indicate a contribution of increased microglial activation and neuroinflammation [23]. In CAD, watershed NAWM shows evidence of lower myelin content, which is associated with poorer cognitive performance, highlighting the vulnerability of NAWM to vascular dysfunction and the need for preventative interventions to preserve brain health in CAD. Future studies should use X-separation technique to help disentangle the effects of demyelination and iron deposition.
Ali REZAEI (Montreal, Canada), Zacharie POTVIN-JUTRAS, Stefanie A. TREMBLAY, Dalia SABRA, Safa SANAMI, Julia HUCK, Brittany INTZANDT, Christine GAGNON, Lindsay WRIGHT, Ilana R. LEPPERT, Christine L. TARDIF, Christopher J. STEELE, Josep IGLESIES-GRAU, Anil NIGAM, Louis BHERER, Claudine J. GAUTHIER
14:24 - 14:26
#47616 - PG163 Personalized DTI-Based Z-Score Analysis of Cerebellar Injury in Mild Traumatic Brain Injury (mTBI).
PG163 Personalized DTI-Based Z-Score Analysis of Cerebellar Injury in Mild Traumatic Brain Injury (mTBI).
Mild traumatic brain injury (mTBI) affects a wide range of individuals and continues to lack reliable objective biomarkers, complicating diagnosis and treatment [1]. Although emerging evidence points to cerebellar involvement in mTBI, this region remains under-investigated due to imaging challenges [2]. Leveraging diffusion tensor imaging (DTI), this study applies a personalized Z-score approach supported by healthy ‘Big Data’ to detect subtle cerebellar microstructural abnormalities in mTBI patients.
This study included 51 symptomatic mTBI patients consisting of 25 females (age: 40.5 ± 11.7 years) and 26 males (age: 45.0±14.1 years). Clinical data including time since injury (TSI) and Post-Concussion Symptom Scale (PCSS) were collected [3]. Each patient was compared to a large, curated dataset of age, sex, and vendor matched healthy controls from multiple open-source repositories [4,5,6]. The healthy control dataset included an equal representation of males and females aged 20 to 65, with Age further divided into nine five-year intervals, beginning with 20-25 and ending with 61-65. Patients were scanned on a GE HealthCare Discovery MR750 3T scanner (system software ver. 29.1) using a 32-channel RF receiver coil. Axial DTI was acquired using a dual spin echo EPI sequence (60 non-coplanar directions, 6 b = 0s/mm2 images, TE/TR = 87/8800ms, b = 1000s/mm2, 2mm isotropic voxels). All healthy controls were scanned on GE 3T machines with varying parameters but adhering to a minimum of 30 diffusion directions and b = 1000 s/mm2. DTI data were preprocessed using FSL tools and registered to MNI space [7,8,9,10]. Scalar maps of fractional anisotropy (FA) and mean diffusivity (MD) were extracted, and 28 cerebellar regions of interest (ROIs) were segmented using a standardized cerebellar atlas [11,12]. A custom pipeline was developed to compute Z-scores comparing each patient to their matched control distribution (by age, sex, and scanner vendor). Abnormality was defined as |Z| > 1.96. Statistical analysis included ANOVAs and linear regression models to assess effects of age, sex, TSI, and ROI. Advanced models like hierarchical clustering and random forests, explored clinical relationships between Z-scores and PCSS. Feature importance and model fit metrics (R², RMSE, η²) were used to evaluate predictors of clinical severity and regional vulnerability. This study presents a personalized cerebellar assessment of mTBI, highlighting the heterogeneity of injury across patients (Figure 1). Case studies revealed distinct regional abnormalities in FA and MD Z-scores, underscoring the limitations of group-level analyses. Among 51 mTBI patients, 41 showed significant Z-score deviations (|Z| > 1.96) in at least one cerebellar region, with sex and age related trends emerging: males had more abnormal MD, females more abnormal FA, and older individuals (>55) showed more widespread abnormalities. ANOVAs and regressions showed age and ROI as significant covariates in FA and MD variability, while sex had a strong effect on symptom severity (PCSS), corroborated by random forest models (Figure 2). Hierarchical clustering revealed distinct patterns by Sex, while feature importance analyses highlighted Age as key for Z-scores and Sex for PCSS. ROI-specific analysis identified left V (FA) and left Crus II (MD) as key regions, though results varied across models. Symptom clustering and modeling further revealed balance problems and sleep issues as top contributors to Z-score variance due to cerebellar abnormalities (Figure 3). Lastly, PCSS had low predictive value compared to Z-scores, suggesting PCSS is limited by subjective bias (Figure 4). Overall, findings emphasize the need for personalized, objective mTBI assessments that consider age, sex, and regional variability. This study highlights the importance of individualized neuroimaging analysis in mTBI patients using DTI metrics, revealing that cerebellar white matter abnormalities are significantly associated with clinical symptoms like balance and sleep disturbances. By accounting for demographics and use of vendor-matched controls, our statistical approaches were able to identify key covariates such as Age, Sex, and time since injury in the personalized assessment of each patient’s injury. Findings support the cerebellum’s role in symptom modulation post-mTBI. We demonstrated the feasibility of a personalized DTI-based Z-score approach to detect cerebellar injury in mTBI, highlighting the variability of microstructural abnormalities across age, sex, and other factors. Findings underscore the importance of individualized assessments and personalized treatment strategies. Cerebellar dysfunction may influence symptom expression, and integrating advanced analytical methods could identify imaging biomarkers. While limitations exist, future work should focus on longitudinal, multi-modal, and AI-driven approaches to enhance diagnostic accuracy and personalize treatment.
Nicholas SIMARD (Hamilton, Canada), Michael D. NOSEWORTHY
14:26 - 14:28
#47928 - PG164 Differences in functional connectivity relate to fine motor recovery during chronic phase post-stroke.
PG164 Differences in functional connectivity relate to fine motor recovery during chronic phase post-stroke.
Stroke is a leading cause of death and long-term disability, with 60% of survivors experiencing lasting impairments [1], particularly in fine motor function [2]. Although rehabilitation often targets these deficits in later recovery stages [3], the neural mechanisms driving fine motor improvement are not well understood. Neuroplasticity, particularly changes in functional connectivity within the motor pathway [4], is thought to play a key role, yet its link to fine motor outcomes remains underexplored. This study aims to address this gap by examining whether resting-state connectivity in the motor pathway differs between patients with and without fine motor recovery post-stroke.
We analyzed data from 42 individuals (informed consent, 27 males; mean age: 58 ± 12 years) with first-time unilateral stroke enrolled in the BSTARS clinical trial [5]. Clinical and neuroimaging assessments were conducted at 5 weeks, 3 months, 6 months, and 12 months post-stroke. For this study, we included 35 scans from the 6- or 12-month timepoints to evaluate chronic-phase recovery. To align with previous research methodologies and validate the classification, gross motor function was first assessed using the Fugl-Meyer (FM) scale, with “Improvers” (n=26) defined by a ≥20-point gain. The primary analysis focused on fine motor recovery, measured by the Nine-Hole Peg Test (NHPT) [6], where “Improvers” (n=14) showed a ≥10-second reduction. Resting-state fMRI was acquired on a Philips 3T scanner with a 32-channel coil (TR = 1.0 s, TE = 25.0 ms, flip angle = 65°, voxel size = 2.3 × 2.3 × 2.5 mm³, multiband factor = 3, duration = 8.2 min). Data were preprocessed using fMRIPrep [7] (realignment, normalization to MNI space—a standard 3D brain reference system—, spatial smoothing, and noise correction), with all lesions mirrored to the right hemisphere. Functional connectivity was analyzed using the CONN toolbox [8] with first-level ROI-to-ROI correlations within motor-related regions: primary motor cortex, premotor cortex, basal ganglia, thalamus, cerebellum, and brainstem [9–16]. Group-level differences were tested using a General Linear Model (GLM) with FDR correction (p-FDR < 0.05). Figure 1 shows the significant differences in connections found between Improvers and Non-improvers of the FM-test. It demonstrates a significant cluster (p=0.0013) with increased connections between the brain stem and the ipsilesional (IL) and contralesional (CL) posterior central gyrus (PostCG), CL and IL precentral gyrus (PreCG) and CL supplementary motor area (SMA). Also Vermis 6 and 7 had an increased connection in Improvers with the IL PostCG and the CL Cerebellum 6 and 8 both had increased connection with the IL PostCG and SMA.
Figure 2 shows the significant differences in connections found between Improvers and Non-improvers of the NHPT. Three significant clusters were found: decreased connections between Cerebellum7 with the motor cortex (p = 0.048), increased connectivity within the frontoparietal network (p=0.049) and decreased connectivity between the premotor network and the frontoparietal network (p =0.049). These findings highlight distinct connectivity differences between Improvers and Non-Improvers. FM-based classification aligns with prior studies, showing altered cortico-cerebellar connectivity [17], brainstem involvement [18,19], and increased connectivity in key motor regions such as the PreCG, PostCG (M1 and S1), and SMA [20]. Focusing on fine motor recovery revealed three key clusters. First, Improvers showed decreased connectivity between cerebellum 7 and the motor cortex (SMA, M1, S1) bilaterally, suggesting reduced integration between these regions. Second, connectivity was reduced between the motor cortex and frontoparietal regions (MidFG, SFG, AG, PaCiG). Each of these has known relevance to motor function: the SFG is linked to complex motor control [21], AG is embedded in key sensorimotor pathways [22], PaCiG seems to interact with the prefrontal cortex to mediate cognitive performance [23], and MidFG is involved in motor planning in a response to sounds [24]. A theory that may explain the results in Figure 2 is that the decreased connectivity in the frontoparietal regions responsible for planning, mediating, connecting and controlling motor function results in a overcompensation, or hyperconnectivity, by the primary motor cortex and cerebellum, which has been speculated before in other neurological disruption studies [25]. Our results support the notion that functional connectivity differs between individuals who recover fine motor skills and those who do not. Specifically, we observed decreased connectivity between the motor cortex and both cerebellum 7 and the frontoparietal network, alongside increased intra-frontoparietal connectivity. These findings may support the theory that hyperconnectivity is a fundamental response to disruption and underscore the frontoparietal network’s role in motor recovery.
Quinten DECKERS (Utrecht, The Netherlands), Jord VINK, Eline VAN LIESHOUT, Bart VAN DER WORP, Johanna VISSER-MEILY, Rick DIJKHUIZEN, Alex BHOGAL
14:28 - 14:30
#47956 - PG165 Decreased levels of N-Acetylaspartyglutamate (NAAG), myo-Inositol (mI), and syllo-Inositol (sI), in cortical brain regions of women exposed to Adverse Childhood Experiences.
PG165 Decreased levels of N-Acetylaspartyglutamate (NAAG), myo-Inositol (mI), and syllo-Inositol (sI), in cortical brain regions of women exposed to Adverse Childhood Experiences.
Adverse Childhood Experiences (ACEs), including abuse, neglect, and maltreatment, are linked to long-term health risks, reduced life expectancy, and changes in brain structure and function [PMIDs: 9635069, 20547282]. Brain imaging studies associate ACE with reduced connectivity between limbic and cortical regions, especially in individuals with mood disorders. Adults with ACE histories show lower prefrontal and hippocampal volumes and altered white matter, though amygdala findings remain mixed [PMIDs: 31445966, 34812558]. Magnetic Resonance Spectroscopy (MRS), particularly the HERCULES method [PMID: 30296560], enables detection of key brain metabolites, offering insight into neurochemical changes related to ACE. However, a distinct neurochemical signature for ACE has yet to be established. Meanwhile, machine learning methods like Logistic Regression (Logit) Support Vector Machines (SVM) and Random Forest (RF) can classify clinical outcomes [PMID: 33980906] i.e, ACE exposure based on data i.e, MRS quantifications . This cross-sectional study aimed to detect ACE-related neurochemical patterns by integrating MRS data with self-reported ACE exposure, supporting non-invasive early detection and intervention strategies to reduce long-term impact.
A total of 43 women aged 19–39 participated in this MRS study, comprising 18 with Low-ACE and 25 with High-ACE exposure. Women were selected due to the higher prevalence [PMID: 40287696] of ACE and the potential for early intervention during young adulthood [PMID: 33308369]. ACE levels were measured using the validated MACE questionnaire [PMID: 25714856], with scores above zero indicating High-ACE. Exclusion criteria included neurological conditions, chronic illness, implants, substance use, pregnancy, or severe depression, the latter screened using the Patient Health Questionnaire-9 (PHQ-9) [PMID: 36865076]. MRS data were acquired using a 3T Philips scanner from the ACC and PFC (via HERCULES) and the hippocampus (via PRESS sequence). Spectral data were analyzed using the Osprey toolbox [PMID: 32603810] and normalized to total creatine (tCr). Group differences in metabolite levels were assessed using Kruskal-Wallis tests, K-W, (p < 0.1, Bonferroni-corrected). To classify ACE groups based on metabolite profiles, machine learning models, including Logistic Regression, RF, and SVM were applied. Women aged 19–31 were grouped based on MACE scores into Low-ACE (–2.46 to –0.8) and High-ACE (0.78 to 2.76). PHQ-9 scores indicated minimal to moderate depression: 1.18–6.62 in Low-ACE and 3.2–13.1 in High-ACE participants. After quality control (tCr FWHM <13 Hz), 42 ACC, 36 PFC, and 27 hippocampus spectra were included. K-W showed significantly lower NAAG in the ACC (p=0.06) and reduced sI in the PFC (p=0.057) among High-ACE participants (Fig.1). Logit identified NAAG, mI, and Glutathione (GSH) in the ACC, and sI, Lactate (Lac), NAAG, and GSH in the PFC as key ACE predictors (AUCs: 0.80 and 0.86). RF confirmed NAAG and mI (ACC) and sI (PFC) as most influential features (AUCs: 0.83 and 0.88), Fig.2. SVM also identified NAAG and mI in the ACC (accuracy = 0.81), sI and GSH in the PFC (accuracy = 0.75), and mI with NAA in the hippocampus (accuracy = 0.63), Fig.3. Findings suggest distinct neurochemical signatures linked to High-ACE exposure. This study explores neurochemical differences between High-ACE and Low-ACE women using MRS and machine learning to identify biomarkers linked to ACE exposure. NAAG and sI emerged as key markers, with lower levels in High-ACE participants. NAAG in the ACC and sI in the PFC were supported by both univariate and multivariate analyses. Reduced NAAG may impact glutamate modulation, neuroprotection, and emotion regulation , while lower sI suggests glial dysfunction. mI also showed non-linear links with ACE in ML models. Despite low depressive symptoms, neurochemical differences persisted in High-ACE women. Hippocampal models had lower predictive accuracy, likely due to technical limitations. Findings suggest specific MRS-detectable changes linked to ACE, independent of current mood disorders. While limited by sample size and generalizability, this study highlights potential for early identification of ACE-related brain alterations and calls for further research using longitudinal designs.[PMIDs: 35163193,22457889,15953489] This study combines MRS and machine learning to examine the neurochemical effects of ACEs in young women. NAAG, mI, and sI emerged as potential biomarkers, with NAAG and sI negatively linked to high-ACE levels in the ACC and PFC. mI showed a non-linear pattern but was consistently selected by machine learning techniques, suggesting relevance. This research offers an important step toward identifying neurochemical signatures of ACE, even in healthy adult populations. Future studies should validate these results in broader populations using longitudinal methods to support early detection and tailored interventions.
Rocio ARTIGAS (Santiago, Chile), Cristián MONTALBA, Claudio PEÑAFIEL, Rodrigo FIGUEROA, Sergio RUIZ, Pablo IRARRAZAVAL
14:30 - 14:32
#47934 - PG166 Neuroinflammatory profiling of high-fat diet effects in IL-1R1KO mice: Insights from multiparametric MRI and indirect calorimetry.
PG166 Neuroinflammatory profiling of high-fat diet effects in IL-1R1KO mice: Insights from multiparametric MRI and indirect calorimetry.
Obesity is a pathological condition with increasing prevalence in our society, due to the complex interplay of biological, socioeconomic and behavioural factors, and is associated with various comorbidities [1]. Chronic inflammation is strongly linked to obesity. Moreover, high-fat diets (HFD) activate pro-inflammatory cascades in the brain, as saturated fatty acids can cross the blood-brain barrier (BBB) and induce pro-inflammatory gene expression [2]. In mice, HFD has been shown to trigger hippocampal dysfunction linked to BBB disruption and neuroinflammation (NI), as well as progressive synaptic and metabolic impairment [3]. Interleukin-1-receptor 1 (IL-1R1), a pro-inflammatory mediator, connects metabolic and inflammatory systems, as its deletion prevents insulin resistance in a diet-induced obesity (DIO) mouse model [4]. Here, our objective is to characterize NI via in vivo multiparametric MRI in IL-1R1KO and wild-type (WT) mice, both fed with standard diet (SD) or HFD. Additionally, we phenotypically assess mice through indirect calorimetry
Male C57BL/6J WT (n=18) and male IL-1R1KO mice (n=19) of 7-8 weeks of age were divided into 2 groups: fed with HFD (n=17) and with SD (n=20) for 20 weeks. In the 10th and 20th week, multiparametric MRI studies were conducted in a Bruker Biospec 7T scanner. Magnetization transfer ratio (MTR) studies, diffusion tensor imaging (DTI), T2 and T2* maps were acquired. Parametric images were generated with a Python-based software and 4 brain regions of interest (ROIs)—cortex (Cx), hippocampus (HPC), thalamus (Thal), and hypothalamus (HTH)—were quantified using ImageJ. Moreover, 5 days after the MRI studies, a phenotyping system (PhenoMaster) was used to analyse mice from each group (n=5-6) during 72 hours (12 hours’ light/darkness cycle), which provided data on indirect calorimetry, locomotor activity and food intake, among other parameters. Linear mixed-effects models were applied to assess the impact of diet and genotype (WT or KO) across ROIs for MRI parameters and an ANOVA analysis was used to evaluate phenotypic differences between groups. MRI studies have detected significantly higher values of fractional anisotropy (FA), in HFD-fed WT mice than in HFD-fed IL-1R1KO mice after 20 weeks. Also, we can see a significantly decrease of FA values of WT obese mice in comparison to WT mice fed with SD, after 10 weeks. While at 20 weeks, the effect of the diet is no longer statistically significant (Figure 1).
We detect significantly higher T2 values, independent of diet, after 10 weeks in every KO mice group and in every area except for Cx and Thal of HFD-fed mice, and in HTH of SD-diet mice. Moreover, after 20 weeks of diet diversification, these differences between genotypes are statistically stronger in every area except for Thal (Figure 2).
In the T2*evaluation, the values are significantly higher in the Cx in HFD-fed WT mice than in the SD-fed WT mice. On the other hand, we observe a difference between genotypes-significantly higher values in HPC and Thal of WT mice- in both diets. This effect of the genotype is present in 10-weeks and 20-weeks mice, only in the HPC. Moreover, we observe an increase in T2* values of KO mice in comparison with WT mice, after 10 weeks with SD (Figure 3).
WT mice gain more weight and faster than KO mice, and both genotype groups show a loss of circadian oscillations in the respiratory exchange ratio (RER) after 10 weeks on HFD and after 20 weeks too. Finally, RER in HFD-fed mice is close to 0.7 at 10- and 20-weeks’ group (Figure 4) From multiparametric MRI studies, the effect of the diet can be detected in FA and T2* values, it is probably due to the effect of NI caused by HFD, vasogenic edema and microstructural damage, respectively.
We observe significant differences between genotypes: The absence of IL-1R-1 seems to decrease FA values after 20 weeks suggesting a progressive damage in these mice. Higher T2 values and lower T2* values in KO mice can suggest NI, caused by vasogenic edema. The absence of Il1r1 gene in mice alters the neuroinflammatory response [5] and this might affect the homeostasis of the brain tissue.
Regarding phenotyping analysis, the higher increase in body weight confirms the acquisition of the obese phenotype in HFD animals. Circadian oscillations in RER are lost in these mice because they are catabolizing mainly fatty acids all day long (RER ~0.7). While SD-fed mice obtain energy from fatty acids, during the hours when they are less active (7 light hours), and from carbohydrates when they are awake, and they can actively eat (7 darkness hours) [6].
To fully interpret the MRI parameter results, we are working in the histology assays and metabolomic post-mortem studies. These findings support the hypothesis that IL-1R1 plays a key role in NI due to HFD-induced obesity and its suppression alters this response. Multiparametric MRI reveals distinct neuroimaging signatures between genotypes and dietary conditions.
Darwin CÓRDOVA-ASCURRA (Madrid, Spain), Raquel GONZÁLEZ-ALDAY, Lidia ESTEBAN-MERAYO, Nuria ARIAS-RAMOS, Jesús PACHECO-TORRES, Pilar LÓPEZ-LARRUBIA
14:32 - 14:34
#47784 - PG167 Time resolved monitoring of brain activity in response to chemogenetic C-Low Threshold Mechanoreceptor stimulation.
PG167 Time resolved monitoring of brain activity in response to chemogenetic C-Low Threshold Mechanoreceptor stimulation.
The C-Low Threshold Mechanoreceptors (C-LTMR) (or C-tactile fibers) are somatosensory neurons that convey gentle affective touch. They modulate pain transmission via spinal inhibitory interneurons and are therefore a potential therapeutic target [1]. In genetically engineered mice [2], the C-LTMR can be selectively activated by pharmacologic agents. This chemogenetic approach in models of chronic pain may enable us to test the hypothesis that the activation of C-LTMR reduces the activity of brain structures involved in pain processing and perception [3]. In this pilot study in non-pain induced mice, we aim to assess whether functional MRI is sensitive enough to map the cerebral projections of chemogenetically activated C-LTMR under anesthesia.
A mouse model with chemogenetically activable C-LTMR based on intersectional genetics (Nav1.8IresFlpo, called Cre+) was explored under isoflurane anesthesia (1-1.6%) in a PharmaScan 70/16 US equipped with 760 mT/m gradients (BGA 9S HP), a volume resonator (72 mm diameter) for emission, a 2x2 elements cryogenic mouse head surface coil for reception, and ParaVison 6.0 software (Bruker). During imaging, rectal temperature was between 35 and 37°C, and breathing rate between 80 and 150 breaths/min. A first cohort (15 ♀ (9 Cre+) and 11 ♂ (5 Cre+) mice) was imaged using a cerebral blood volume weighted (CBVw) approach before and for 85 min after intravenous (iv) injection of an USPIO (MoldayION, BioPAL) (Fig 1a). A 20-minute interval after USPIO administration was used for CBVw signal detrending (to account for continuous USPIO washout) before Clozapine N-oxide (CNO), a C-LTMR activating drug, was injected intraperitoneally (ip) and CBVw signal was acquired for another 65 minutes. In addition, 4 experiments (without MRI) were performed without USPIO, and one without CNO to study pharmacological interactions. A second cohort (17 ♀ ( 7 Cre+) and 14 ♂ ( 7 Cre+) mice) was imaged with a pseudo continuous arterial spin labeling (pCASL) sequence [4] repeated over 60 min to assess cerebral blood flow (CBF) changes (Fig 1b). C21 was used as C-LTMR activation drug [5]. Cre- mice without activable C-LTMR represented the control group. After motion correction with FSL, images were processed with ImageJ for CBVw mapping (Fig 1a) [6], and with MP3 for CBF mapping [4,7] (Fig 1b) without spatial but with temporal smoothing (5 min for the CBV change, 4 min for the CBF change). Maps of CBV and CBF changes were analyzed individually, with particular attention to ROIs in the somatosensory cortex and brain structures related to emotion processing. The CBVw approach was successfully carried out in 9 ♀ (5 Cre+) and 7 ♂ (4 Cre+) mice. Other acquisitions were excluded due to technical problems, uncorrectable head motion artifacts, unreliable USPIO injections and respiratory distress which was observed in 10/26 mice 5 to 25 min after CNO administration. A CBV increase was observed in the insular cortex of 3 mice without respiratory distress starting 10 to 25 min after CNO administration (Fig 2). These 3 mice were all ♀ Cre+, and showed a peak CBV change of 15 to 25 % 45 to 55 min after CNO, which agrees with the known pharmacokinetic profile of CNO, and a sexually dimorphic behavior observed in pharmacologic studies. All other mice, including 2 ♀ Cre+ mice, had no reliable CBV change. No vasoactive effect of CNO was noticed in extracerebral tissue. Respiratory depression occurred also when CNO alone was administered to anesthetized mice, excluding an adverse effect of the USPIO. The CBF approach was successfully carried out in all mice of the second cohort, without any adverse effect of the C21 compound. However, no reliable CBF change was observed in the insular cortex or any other brain structure or extracerebral tissue (Fig 3). Individual variability was observed in the first cohort with respect to the respiratory depression and hemodynamic effect in insular cortex upon CNO administration. Despite possible off-target effects of CNO or its metabolites, identification of C-LTMR specific effects are expected since cerebral hemodynamic effects did not occur in the control group (Cre-) without activable C-LTMR.
Although being sensitive and showing results agreeing with expectations, the CBV approach was considered technically challenging, invasive (iv access), and prone to confounding effects (USPIO washout). We therefore attempted to reproduce the results with a non-invasive time resolved functional MRI technique and C21, a drug known to have reduced off-target effects [5]. Despite this quantitative CBF approach, no C-LTMR specific hemodynamic effect has been observed so far. Group-wise comparisons are still ongoing. A CBVw MRI approach requiring iv injection of an USPIO showed cortical CBV changes upon chemogenetic stimulation of C-LTMR compatible with the known pharmacokinetics of CNO. However, these were not yet reproduced using a non-invasive time resolved quantitative CBF mapping technique.
Khouloud BENZZAOUIA (Marseille), Guillaume ROBERT, Anaëlle MÉAULLE, Ludivine GUYOT, Isabelle VALET, Abdelaziz MOQRICH, Angèle VIOLA, Adriana Teodora PERLES-BARBACARU
14:34 - 14:36
#47598 - PG168 Multi-modality data integration of electrocorticography and fMRI for investigating cross-scale brain network organization in patients with brain tumors.
PG168 Multi-modality data integration of electrocorticography and fMRI for investigating cross-scale brain network organization in patients with brain tumors.
The functional organization of the brain spans multiple spatial and temporal scales. In glioma patients, tumor-neural interactions disrupt brain network architecture at the microenvironment level, locally near the tumor, and globally across the brain (1–4). These disruptions alter functional connectivity, and our understanding of these processes is limited (5). For effective surgical planning, individualized functional mapping is essential to identify and preserve critical regions and minimize postoperative deficits (6). After tumor resection, the brain often undergoes reorganization accompanied by cognitive changes, but the mechanisms remain unclear.
In this study, we examine how tumors affect functional organization of the hemisphere they inhabit compared to the contralateral hemisphere. We focus on regions associated with executive functions (e.g., attention, cognitive flexibility, task-switching), and examine the frontoparietal network (FPN) which supports these higher-order cognitive processes. To that end, we developed a cross-modality pipeline for integrating data from two neuroimaging modalities: electrocorticography (ECoG) and resting-state functional MRI (rs-fMRI). ECoG captures local meso-scale neural activity from the cortical surface with uniquely high spatiotemporal resolution. rs-fMRI is used to characterize functional connectivity at the whole-brain scale. In our prior work we showed that tumor-infiltrated cortex participates in large-scale cognitive circuits (7). Here we expand this work to investigate healthy-appearing regions and laterality of connectivity patterns.
Data from 11 glioma patients were analyzed to assess local involvement in executive function and large-scale functional connectivity. ECoG and longitudinal rs-fMRI data were collected for each patient. ECoG was recorded from the frontal cortex during awake surgery while patients performed two counting tasks with varying cognitive demand to identify regions engaged in executive function (8). For each patient, 4–12 electrodes were placed on the tumor hemisphere (sampling rate: 10 kHz). Following preprocessing (re-referencing, line-noise filtering, down-sampling), changes in high gamma power (70-250 Hz) were computed per electrode, indicating local task-related activity. rs-fMRI BOLD data were collected before and at 3 months after surgery (Siemens Magnetom Prisma-fit 3T scanner, TR=1060 ms, TE=30 ms, 2 mm isotropic resolution, FOV=192x192 mm2, acquisition time: 9 min and 10 s) and pre-processed using Independent Component Analysis to remove noise. Seed-based connectivity maps were generated by correlating the fMRI time series of a 2.5 mm radius sphere around each electrode location with all other brain voxels, capturing individual whole-brain connectivity patterns (Fig. 1). Median connectivity with canonical resting-state networks was computed for each electrode (9). Electrodes on both healthy-appearing and tumor-infiltrated cortex showed task-related high gamma activity, indicating engagement in executive function. Before surgery, whole-brain connectivity with canonical networks was similar for electrodes on healthy-appearing and tumor-infiltrated cortex. Hemispheric differences were observed when examining connectivity with each hemisphere separately. Functional connectivity with the FPN, where 62% (33/53) of the electrodes placed on healthy-appearing cortex were located, was significantly larger in the tumor hemisphere than the contralateral side (Fig. 2A). This asymmetry was primarily driven by electrodes that were task-responsive or located within the FPN (Fig. 3). Postoperatively, this hemispheric difference in FPN connectivity was no longer present (Fig. 2B; mixed linear ANOVA model, interaction between time and hemisphere, p<0.05). Our findings demonstrate that gliomas alter functional connectivity patterns, and that tumor resection leads to reorganization of these patterns at the whole-brain level. We observed asymmetry in connectivity preoperatively, particularly with the FPN, which was absent postoperatively, suggesting network reorganization. The preoperative asymmetry may reflect either compensatory processes or abnormal disruption that occur during tumor growth. Glioma patients often exhibit cognitive impairments (10), and further research is needed to shed light on how alterations in network connectivity relate to behavior and cognitive outcomes following surgery. This study highlights the value of cross-modality integration of ECoG and fMRI to map functional networks in glioma patients and advance our understanding of brain reorganization caused by tumors and their removal. Our approach and findings may lead to the development of patient-tailored treatment and rehabilitation strategies.
Chemda WIENER, Ayan MANDAL, Moataz ASSEM, Rafael ROMERO-GARCIA, Pedro COELHO, Alexa MCDONALD, Emma WOODBERRY, Michael HART, Stephen PRICE, Robert MORRIS, John SUCKLING, John DUNCAN, Thomas SANTARIUS, Yaara EREZ (Ramat-Gan, Israel)
|
Salle Major |
|
"Friday 10 October"
Added to your list of favorites
Deleted from your list of favorites
D24
14:00 - 15:30
FT1 LT - Hardware technology
FT1: Cycle of Technology
14:00 - 14:02
#45680 - PG169 A unipolar high-performance head gradient for high-field MRI without encoding ambiguity.
PG169 A unipolar high-performance head gradient for high-field MRI without encoding ambiguity.
Gradient coils for the z-axis traditionally follow the principle of a Maxwell pair, comprising two sections that generate field of the same spatial structure but opposite polarity [1,2]. The two field lobes superimpose to form a bipolar field with a zero in the iso-centre and a surrounding linear range (Fig. 1B). Outside this range the field reaches maximum excursions, beyond which it gradually drops back to zero, causing ambiguity of gradient encoding. To prevent the related backfolding, the unambiguous range must be made sufficiently long [1]. However, this comes at great expense in terms of gradient performance [2,3]. Therefore, the range required to prevent backfolding is commonly contained by limiting the spatial coverage of RF transmission and detection [4].
While long-established for clinical whole-body systems, this approach to gradient ambiguity is less favourable for cutting-edge brain imaging, which increasingly relies on field strengths of 7T and beyond, where RF fields are less contained. At the same time, advanced neuro-MRI demands ever-higher gradient performance, which is increasingly implemented through head-only gradients [5-23]. These, in turn have intrinsically smaller unambiguous range, and high-field imaging with head gradients has indeed been reported to suffer from backfolding [24-27].
To address this issue, the present work demonstrates an alternative approach to gradient ambiguity. It takes advantage of the fact that, when imaging the head, ambiguity is of concern only on one side of the imaging volume. Gradient encoding without any ambiguity can thus be performed with a unipolar rather than bipolar z-gradient field, effectively reducing the Maxwell pair to one of its halves (Fig. 1C) [28]. This concept is implemented for a high-performance head gradient at 7T. The absence of any backfolding is verified experimentally.
A head gradient with unipolar design was developed for a Philips 7T system (Fig. 2A). It is operated with the available standard environment, including a dual-mode amplifier (Copley 787). A free bore diameter of 39 cm accommodates typical head coils and a conical opening provides appropriate patient access [17]. In the linearity volume (LV) of 22x22x20 cm3 the deviation in gradient strength is < 20%. Maximum gradient strength and slew rate are 200 mT/m and 560 T/m/s.
Optimisation of a z-gradient with unipolar design within the given constraints led to the field shown in Fig. 1C. As a side effect, the design introduces an additional field offset in the imaging volume. For the xy-dimensions a conventional bipolar design was employed. To contain current density, a system of double layers of conductors was chosen. Cooling is based on a combination of hollow and solid conductors. Figs. 2B-C show the completed gradient.
The software of the scanner was modified to address the field offset associated with the unipolar design by an equivalent off-centre in the z-dimension, corresponding to modulation and demodulation of RF signals. In addition, preparation procedures, image reconstruction, and image processing were adapted accordingly.
To investigate the ambiguity issue, imaging was performed with a large FOV in phantoms and in vivo using an RF transmit-receive quadrature birdcage head coil (Nova Medical). Gradient non-linearity (NL) correction was applied based on calculated field maps [29]. Imaging with the unipolar gradient was successfully performed. The phantom and in vivo experiments in Figs. 3 and 4 demonstrate that with the unipolar gradient design no backfolding occurs from the neck and trunk region into the LV. Relative to conventional gradients, one practical consequence of the unipolar design is the need to consider the introduced field offset during different parts of the MR procedure. The efforts and implications of these changes will strongly depend on the structure, implementation, and accessibility of the software of the particular MRI scanner.
Another consequence is greater maximum field strength, created in the signal-free range. Somewhat counter-intuitively it was found, that the higher field maximum does not prevent clearly competitive performance with the bipolar design.
Moreover, the higher maximum field may raise concerns regarding PNS. That is offset, however, by the fact that the maximum occurs only on one side and well outside the subject. Nevertheless, PNS is potentially increased at the top of the head. For the neck and shoulders, the unipolar approach is expected to reduce PNS. Initial testing indicated a rather benign PNS behaviour. A unipolar gradient design was demonstrated that for head gradients fundamentally solves the ambiguity issue of the commonly used bipolar fields. Thus, gradient and RF coil development are disentangled. The prospect of gradient systems based on this sort of design holds promise for all advanced neuroimaging that demands high gradient performance, and will make the greatest difference at 7T and beyond.
Markus WEIGER (Zurich, Switzerland), Overweg JOHAN, Franciszek HENNEL, Emily Louise BAADSVIK, Samuel BIANCHI, Björkqvist OSKAR, Roger LUECHINGER, Metzger JENS, Michael ERIC, Thomas SCHMID, Lauro SINGENBERGER, Urs STURZENEGGER, Erik OSKAM, Gerrit VISSERS, Jos KOONEN, Wout SCHUTH, Jeroen KOELEMAN, Martino BORGO, Klaas Paul PRUESSMANN
14:02 - 14:04
#47897 - PG170 Development of LNA for multichannel coil array in low-field MRI.
PG170 Development of LNA for multichannel coil array in low-field MRI.
Development of modern low-field MRI systems depends on efficient high-quality electronics to achieve a high signal-to-noise ratio (SNR) in the resulting image. First stage of the MR-scanner receive chain - low-noise amplifier (LNA) - has a significant impact on received signal SNR. LNAs existing on a market are available for the frequencies down to 19 MHz [1]. The closest specific amplifier for low-field MRI were described in papers [2,3], where authors demonstrated a 32 MHz LNA. Attention to lower frequency ranges has grown with the recent raise of interest to low-field MRI systems including portable configurations [4,5]. Recent hardware advances improved image quality making imaging at lower field strength clinically relevant and feasible [6]. In work [7] it was shown that using a low input impedance amplifier allows to decrease mutual coupling between coil elements of an array. In this work we aimed to develop a simple and low-cost LNA with low input impedance that could be used for 0.5T (21.2 MHz) and 0.07T (3MHz) low-field systems.
The computer-aided engineering CAE simulation software was used to numerically optimize the design of an LNA. The amplifier circuit consists of two stages based on the BFR193F [8] bipolar junction transistor (BJT). First stage of the amplifier contains BJT in a common-base (CB) configuration, which provides low input impedance and ensures a 50 Ohm impedance match at the output. The second stage of the amplifier is implemented using the same BJT transistor in a common-emitter (CE) circuit, ensuring sufficient gain and 50 Ohm matching both at the input and output. To enhance the stability of the common-base circuit a resistor was added in series with radiofrequency (RF) choke. For the second stage resistive feedback and inductive degeneration were applied to achieve unconditional stability of the amplifier. By numerical simulations of 3 MHz (Fig.1) and 21.20 MHz amplifier circuits (Fig. 2) S-parameters, noise figure (NF) and stability characteristics K of the amplifier were calculated. For experimental evaluations, a prototype of the 0.50 T (21.20 MHz) amplifier was assembled to verify numerical results. The vector network analyzer (VNA) OBZOR TR1300/1 was used for S-parameters measurements and LNA adjustment. Noise figure measurements were performed with Agilent N8973A noise figure analyzer. As a result of numerical simulations, two amplifier configurations operating at 3 MHz and 21.20 MHz were obtained. According to the results of numerical simulations the 3 MHz LNA has 53.30 dB gain, 5 Ohm input impedance, -25.70 dB reflection coefficient at the output, 0.84 dB noise figure (Fig. 3), 20 dBm input compression point (P1dB) and K-factor greater than 22.
The 21.20 MHz LNA has 47.22 dB gain, 5 Ohm input impedance, -39.50 dB reflection coefficient at the output, 0.82 dB noise figure (Fig. 4), 17.78 dBm input compression point (P1dB) and K-factor greater than 14 according to the numerical results.
To validate the results of numerical simulations a prototype of the 21.20 MHz amplifier was assembled. Experimentally obtained S-parameters have slight differences compared to numerical results. The prototype gain is 1 dB lower than predicted due to parasitic parameters of real components. Reflection coefficient is equal to -18.89 dB, difference compared to -39.50 dB in simulation is due to home-made inductance for output impedance matching. The measured value of the noise figure is 0.74 dB (0.08 dB better than in simulation). The measured 21.20 MHz LNA characteristics are in good agreement with numerical data. Compared to the closest commercial WanTcom amplifier (19 MHz) developed 21.20 MHz LNA has higher gain (28 dB for WanTcom vs 42.77 dB for 21.20 MHz). However, commercial amplifier outperforms the proposed design in NF value (0.50 dB for WanTcom vs 0.80 dB for 21.20 MHz) and input impedance (1.80 + 0j Ohm for WanTcom vs 5.20 + 2j Ohm for 21.20 MHz) which improves coil array decoupling. In this work two LNAs based on a common base first stage circuit using low-cost BFR-193 BJT operating at 3 MHz and 21.2 MHz were developed. The prototype of the 21.20 MHz amplifier demonstrates 5.2 + 2j Ohm input impedance, 0.80 dB noise figure and 24 dBm P1db. The numerical model of the 3 MHz amplifier have shown 5 Ohm input impedance, 0.84 dB noise figure and 20 dBm P1dB. Characteristics of the 21.20 MHz amplifier are comparable to modern commercial WanTcom amplifier. The designed amplifiers provide cost-effective solutions for low-field MRI receivers making such scanners more accessible. Within our near future plans is refinement of amplifier prototypes and release of final models as open source project.
Mikhail MURZIN (St. Petersburg, Russia), Aleksei NASONOV, Anna HURSHKAINEN, Georgiy SOLOMAKHA
14:04 - 14:06
#45835 - PG171 Development of a transducer system for MR Elastography at 3T and 7T.
PG171 Development of a transducer system for MR Elastography at 3T and 7T.
Magnetic Resonance Elastography (MRE) is a technique for assessing tissue stiffness [1], used to detect disease-related changes in mechanical properties of tissues, especially in cardiac MRE (cMRE), where myocardial stiffness provides a marker for evaluating heart function[2].
This transducer is featuring a rotating unbalance, driven by a turbine and compressed air, all inherently MR safe, no metals or conducting materials are used. Adding mass by inserting up to six rods, sufficient actuation force can be generated to produce detectable displacements deep in the body.
It is designed to be flat to fit in between the patient and the coil. Increased transducer height extends the distance from the coil to the region of interest, reducing MR signal strength. While prior transducer designs have incorporated flat profiles, variable masses [3], or pneumatic actuation [4], no solution has yet integrated all these features into a single compact device.
The transducer prototype uses the principle of a rotating unbalance to generate vibration. It consists of a 3D printed turbine connected to two unbalances with slots for exchangeable masses. These are placed between four ceramic bearings, which are all placed in a CNC milled case. Plastic connectors guide pressurized air from and to the turbine. A fiber optic sensor is used to measure the rotation frequency of the transducer. The transducer is controlled by a control-unit which consists of a proportional valve, a pressure sensor, a microcontroller and a data-acquisition card (DAQ). A PID controller is constantly updating the valve position to control the rotation speed.
This concept allows to vary frequency and generated vibration force independent from each other. Frequency can be controlled via input pressure and force via the weight configurations.
Also, in-scanner measurements were performed to test both the transducer as well as an in-house modified FLASH with motion encoding gradients (MEG). Scanner measurements were performed at a Siemens (Siemens Healthineers, Erlangen, Germany) 3T (PrismaFIT, spine array and body-18 matrix coils, TR/TE: 60/6ms , FOV: 380 mm, 3 mm voxel isotropic, 8 wave phases) and 7T (MAGNETOM 7T Plus, 2 channel dipoles array (RAPID Biomedical, Rimpar Germany, TR/TE 60/6ms: , FOV: 380 mm, 3 mm voxel isotropic, 12 wave phases).
For all tests an ultrasound gel phantom (Ultragel Medical Kft., Hungary) with a known shear modulus of 0.9 kPa [5] was used. The transducer was set to 50 Hz. Shear stiffness maps were calculated with the k-MDEV inversion method in the BIOQIC [6] elastography tool. All parts of the developed transducer prototype can be seen in Figure 1, as well as the unbalance with variable masses and the control unit.
For performance evaluation the frequency was measured over 15 min of rotation was 49.99 ± 0.24 Hz, showing a very accurate control.
The pipeline where elastograms of the complex shear modulus are depicted can be seen in Figure 3 and Figure 4. This study shows advancements in developing a new MRE transducer. It shows very stable rotation speeds, tested between 20-100 Hz, excellent compatibility in the MR environment, as well as connectivity with the scanner.
The elastograms show similar values in the phantom when compared with known data from literature [5]. The system performed both in 3T and 7T environment. Once IRB approval is obtained, in vivo performance will be evaluated. Here, a functional prototype for a novel MR elastography transducer was presented. Most importantly, it allows independent changes in amplitude and speed and fits between coil and patients thus increasing the flexibility of applications. Its MRE performance was demonstrated both at 3T and 7T.
Lorenz KISS (Vienna, Austria), Christopher BREITENFELDER, Stefan WAMPL, Marcos WOLF, Hodul ANDREAS, Quang NGUYEN, Martin MEYERSPEER, Albrecht Ingo SCHMID
14:06 - 14:08
#46875 - PG172 Investigating magnetic properties of 3D-printable materials at different field strengths using SQUID Magnetometry.
PG172 Investigating magnetic properties of 3D-printable materials at different field strengths using SQUID Magnetometry.
3D printing has become widely adopted across various aspects of MRI research. Its applications range from phantom construction to the manufacturing of gradient coils and other components, particularly in low-field MRI systems.
Previous work that investigated the magnetic properties in the context of MRI has been carried out by placing the sample in an MRI scanner surrounded by deionized water with known magnetic susceptibility and then measuring the field distortion [1-3].
A limitation of this approach is that magnetic susceptibility measurements are confined to the B0 field strength of the MRI scanner being used. While 3D-printing plastics are generally known to be diamagnetic, the presence of contaminants, such as colorants or composite materials, can result in field-dependent magnetic susceptibility. Far below the saturation field strength of ferromagnetic contaminants, the susceptibility can be shifted significantly towards positive values, while above the saturation field strength the diamagnetism of the bulk polymer dominates the magnetic properties of the 3D-printed materials.
To investigate this field-dependent magnetic behavior, SQUID (Superconducting Quantum Interference) magnetometry was used due to the high sensitivity and the ability to vary the field strength when measuring the magnetic moment of the samples.
Samples
The materials were selected to cover a range of materials commonly used in MRI applications (Table 1).
Two different types of 3D-printing techniques were investigated:
- Fused Deposit Modeling (FDM) samples were printed on a large-volume 3D printer (V-Core 4.0 500mm, RatRig, Faro, Portugal) using both a 0.4mm brass nozzle and a 0.4mm hardened steel nozzle for abrasive materials. The infill was set to 100% and the layer height to 0.2mm.
- Additionally, a 3D printable resin sample was produced using a resin 3D printer (LD-002R, Creality, Shenzen, China).
All samples were designed as cylinders with a height of 6mm and a diameter of 5.6mm.
Magnetometry
A SQUID magnetometer (MPMS XL, Quantum Design, San Diego, USA) was used to carry out the measurements.
The samples were mounted in a SQUID magnetometry sample holder and secured using thin cotton strings(Figure 1).
All measurements were conducted at 300K in a temperature-controlled environment.
The hysteresis loop was measured from -3T to 3T, with variable step sizes around the zero transition.
The volume susceptibility was then calculated from the magnetic moment.
The calculated volume susceptibility was the compared to an existing open database of 3D-printable materials (MaDaMEPro) [4]. Figure 2 shows the hysteresis loop of a collection of 3D printed samples with and without ferromagnetic contaminants. Commonly used materials such as PLA red and PETG CF exhibit a field-dependent magnetic susceptibility, while the other samples show a more linear behavior.
Table 2 shows the volume susceptibility of the samples at different field strengths and the deviation from the literature. Significant deviations at lower field strengths can be observed for contaminated samples. SQUID magnetometry revealed the presence of ferromagnetic contaminants within several 3D-printed samples (Figure 2). Of particular interest is the behavior of the red PLA sample, as this material is frequently used in MRI applications.
At B0 field strengths of 50mT, typical for low-field MRI scanners, the bulk behavior of the samples exhibits a positive magnetic susceptibility. This contrasts with the expected diamagnetic behavior of materials commonly used in 3D printing and contradicts existing literature values where diamagnetic behavior observed at higher field strengths is extrapolated to lower B0 field strengths [1].
When comparing susceptibility values with the literature, several factors must be considered. Neither the materials nor the printers used are identical to those referenced in the literature, making direct comparisons of absolute values problematic. However, the relative susceptibility values are comparable and demonstrate clear deviations for samples containing ferromagnetic contaminations at lower field strengths (Table 2).
Future research will investigate whether these significant deviations from water's volume susceptibility can influence image quality at lower MRI field strengths. This will involve printing MRI phantoms using different materials and imaging them on a low-field (50mT) MRI scanner. Additionally simulations will be carried out to observe the effect of different levels of contaminants at different B0 field strengths. Samples that are widely used in MRI exhibit quasi-ferromagnetic behavior at lower field strengths. Further investigation will be carried out to determine if the contaminations might have an effect on the MRI image.
For the moment, the results indicate that caution should be taken when using colored or compound materials in low-field MRI applications.
Julia PFITZER (Graz, Austria), Jakob RATZENBERGER, Marc RUOSS, Martin UECKER, Hermann SCHARFETTER
14:08 - 14:10
#47685 - PG173 Differentiable dipole-based optimization of permanent magnets for homogeneous b0 fields in low-field mri.
PG173 Differentiable dipole-based optimization of permanent magnets for homogeneous b0 fields in low-field mri.
Permanent magnet based Low-field MRI systems have attracted growing interest thanks to their affordability and ongoing performance gains. However, image quality is often limited by inadequate B0 homogeneity, arising from complex, nonlinear interactions among thousands of permanent magnets. Although permanent magnets are low-cost and maintenance-free [1-2], designing such large assemblies poses a high-dimensional optimization and manufacturing challenge. Achieving a high field uniformity needed for diagnostic imaging demands both computationally efficient algorithms and practical manufacturability. Previous approaches—ranging from genetic algorithms tuning discrete magnet parameters [3] to reinforcement-learning heuristics [4]—rely on predefined, discretized design spaces.
In this work, we introduce a fully differentiable framework that treats magnet positions, orientations, and sizes as continuous variables. While our method can optimize all positional and rotational degrees of freedom, we here illustrate its use on angular-orientation optimization. By leveraging gradient-based updates across the entire assembly, our approach achieves finer control of B0 homogeneity and enables more scalable exploration of permanent-magnet designs for low-field MRI.
Each magnet in a permanent magnet-based low-field MRI system can be represented by size, shape, orientation, material properties, and magnetization strength. Multiphysics simulations such as CST [5] or COMSOL [6] use finite-element and boundary-element discretizations to compute magnetic fields accurately, but their reliance on non-differentiable computations complicates direct integration with fast optimization methods.
In our differential optimization framework we make the assumption of relatively small magnets compared to the radial distance from the center of the volume of interest (VOI), allowing us to model each magnet as a single magnetic dipole moment [3,7,8]. Under this model, the Biot-Savart law [9] provides a differentiable, closed-form mapping. In contrast, heuristic methods—genetic algorithms [3] and reinforcement learning [4]—rely on discrete parameter searches that limit design generalisability.
We formulate a continuous optimization to minimize field inhomogeneity within a spherical VOI by tuning each magnet’s in-plane and out-of-plane rotation (Image 3, right). For the results at hand, we utilized the Adam optimizer [10], though any optimizer based on gradient descent is applicable with our proposed approach. We initialized the optimizer with 10 different learning rates ranging from 0.001 to 0.01.
As a baseline comparison, we simulate a handcrafted Halbach-array assembly, consisting of 14 concentric layers of 3 rings (Image 3, left) and designed to generate approximately 50 mT within a 20 cm-diameter VOI for human-head imaging.
We assess performance with two loss metrics: direct homogeneity (min-max variation of the field) and mean squared error (MSE) against a target field. Each optimization executes 1000 iterations, empirically sufficient for convergence. The proposed framework can integrate additional objectives, manufacturing constraints, or alternative geometries for permanent-magnet MRI design. The initial homogeneity of the handcrafted system was 96197 ppm within a 20 cm diameter VOI, with an average field strength of 52.37 mT. After optimization, homogeneity improved to 8588±1495ppm, while the field strength slightly decreased to 44.43 ± 0.63 mT — a 15.16 % reduction. This corresponds to an 11.2-fold improvement in homogeneity. In a separate MSE optimization targeting 50 mT, homogeneity improved to 33281±1907 ppm, with a field strength of 49.989 ± 0.002 mT, yielding a 2.89-fold improvement. All optimizations converged within 1000 iterations. Detailed results are shown in Images 1 and 2. The proposed differentiable optimization framework significantly enhances magnetic field homogeneity in low-field MRI. Applied to our handcrafted Halbach-array baseline, it converged within 1000 iterations to achieve an 11.2-fold homogeneity improvement while maintaining field strengths close to the target range. By treating magnet parameters as continuous variables, our method flexibly optimizes objectives such as maximizing field strength, improving homogeneity, or achieving a uniform target field across the VOI. Moreover, the differentiable formulation enables rapid exploration of alternative system architectures and magnet distributions. To illustrate versatility, we also optimized a handcrafted Moebius-strip configuration (Image 4), demonstrating adaptability to varied geometries. We present a novel, differentiable optimization framework for the design of permanent magnet arrays in low-field MRI. This method allows efficient and accurate optimization of field homogeneity and strength, adaptable to a wide range of initial configurations and system goals. By offering flexibility and robustness, it holds great potential for advancing MRI technologies.
Kostiantyn LAVRONENKO (Aachen, Germany), Marcel OCHSENDORF, Julian THULL, Schulz VOLKMAR
14:10 - 14:12
#46445 - PG174 A novel 64-channel ultra-flexible RF coil for enhanced prostate, rectal and pelvis imaging.
PG174 A novel 64-channel ultra-flexible RF coil for enhanced prostate, rectal and pelvis imaging.
Current RF coils for prostate and pelvic imaging struggle with limited depth sensitivity and coverage, especially across different body sizes. These limitations reduce diagnostic accuracy [1]. Previous solutions, such as endorectal coils, offer proximity to the prostate but compromise patient comfort and do not cover larger regions accurately [2-4]. The 50-channel coil described in [5] has shown improvements, with SNR increased by ~36% with respect to a previous 30-channel model. However, further enhancements are needed not only for a better SNR, but also depth and reliability. This study addresses these challenges by developing a novel 64-channel coil with an optimized design to enhance imaging across a wide range of patient anatomies using AIR Technology [6,7].
We designed an ultra-flexible 64-channel coil for imaging the pelvic, rectal, and prostate areas, accommodating body sizes from the 5th percentile female to the 95th percentile male. The array consists of two identical halves (posterior/anterior) connected at the perineum by an adjustable strip. Each half includes linear and staggered regions with 26 loops (10.6 cm) and 6 loops (14.4 cm), enhancing signal strength and depth. We used flexible AIR Technology loops with critical overlap [8], as well as overlaps ranging from 15% to 30%, enabling optimal decoupling with the help of preamplifiers. A 95th percentile male phantom was modeled in HFSS (Figures 1a and 1b) and the wrap was simulated in Blender for validation (Figures 1c and 1d).
To fabricate the coil array, we first mapped each half on the fabric and proposed a path for the wiring of all the loops. We then sewed the loops using both tack and zigzag stitching, placed the associated electronics and wiring, and added floating cable traps to help filter common-mode noise (Figure 2a). We then tested all the loops in each half and added Nanonomex fabric layers to protect the coil elements (Figure 2b). After that, we sewed the external Dartex layers and incorporated the adjustable strips (Figures 2c and 2d).
Finally, we tested the coil array on a 3.0 T SIGNA Architect Scanner (Hospital Politécnico y Universitario La Fe, Valencia, Spain) using a protocol for the prostate area, to verify loop functionality. Additionally, we conducted thermal tests using a Fluke infrared temperature gun to ensure compliance with temperature standards. Then, we performed phantom scans and the first in vivo tests. The high flexibility of the coil array enables it to conform easily to pelvic anatomy. Phantom imaging (Figures 3a-3c) confirmed that all loops within the array were working. Thermal testing recorded maximum temperatures below 34 °C, remaining well within safety thresholds for both patient-contact components (41 °C) and those handled by technicians (48 °C). Additionally, the noise covariance map for the 64-channel coil is presented in Figure 3d, observing that channel 14 exhibited noise levels significantly higher than the rest. Finally, initial in vivo tests are shown in Figures 4a and 4b. The 64-channel ultra-flexible coil array offers a high degree of anatomical conformity, effectively covering up to the 95th percentile of the male population. Its mechanical flexibility and design improve patient comfort by eliminating the need for complex lateral closures and incorporating an adjustable perineal support, ensuring a close fit across a broad range of body types.In addition, the thermal measurements ensure the coil to remain within clinical safety thresholds.
Phantom scans confirmed that the coil operated correctly overall, with the exception of channel 14, which exhibited excessive noise. A more detailed evaluation will determine whether replacement is necessary. Nevertheless, this loop is located on the lateral side of the array, making its contribution less critical than that of other elements. Aside from this, in vivo scans acquired with the 3.0 T SIGNA Architect Scanner demonstrate promising results in image quality when compared to the current coil, which combines a posterior spine coil and an MP anterior coil. These preliminary findings suggest that the proposed array could offer increased diagnostic capability in prostate, rectal, and pelvic imaging, considering also that the sequence has not been optimized for the new coil yet. Moreover, the data collected will be evaluated soon more in-depth to define coil sensitivity, penetration depth, and overall performance. This study presents a novel, high-density, ultra-flexible RF coil array specifically designed for prostate, rectal, and pelvic imaging. The array demonstrates strong anatomical adaptability and improved patient ergonomics. Early phantom and in vivo imaging results reveal encouraging image quality compared to the standard clinical coils currently in use. These results support the feasibility of implementing this coil in clinical settings and motivate further analysis into its performance metrics and diagnostic benefits.
Jesús CONEJERO (Valencia, Spain), Jana VINCENT, José Miguel ALGARÍN, Edward BAUS, María De La Luz JURADO-GÓMEZ, José DE ARCOS, Arnaud GUIDON, Victor TARACILA, Fraser ROBB, Leonor CERDÁ-ALBERICH, Luis MARTÍ-BONMATÍ, Joseba ALONSO
14:12 - 14:14
#47899 - PG175 A wireless quadrature Rx-only coil based on electromagnetically decoupled Helmholtz resonators for 1.5T MR mammography.
PG175 A wireless quadrature Rx-only coil based on electromagnetically decoupled Helmholtz resonators for 1.5T MR mammography.
Inductively coupled (IC) wireless radiofrequency (RF) coils are intensively studied in the recent years due to customer benefits such as absence of vendor-specific interfaces as well as increased safety performance caused by the absence of cable connections [1-4]. Moreover, IC wireless coils have cost-efficient configurations due to no need of low-noise non-magnetic receive electronics and proprietary connectors. IC wireless coil operational principle is based on inductive coupling with transceiver body coil of MR scanner. Dedicated breast IC wireless coils are of special interest due to the low availability of breast coils in clinical practice. Therefore, several solutions are available to the moment [1,3] showing promising results in the context of MR mammography. These solutions are based on the sets of coupled volume resonators covering the targeted ROI of human breast. Though the results obtained with coupled resonators are interesting, there is still a room for improvements in the context of IC wireless coil receive sensitivity (SNR). Moreover, the known wireless coils for breast imaging operate as transceiver (Tx/Rx) coils and the standard clinical workflow is violated due to the need of manual reference voltage calibration.
In this work, we demonstrate the first receive-only (Rx-only) quadrature wireless coil for bilateral breast imaging at 1.5T. Two pairs of decoupled Helmholz-type resonators are employed having additional benefit as side access to breast tissues. The Rx-only operation is due to the passive decoupling diode circuits. The quadrature coil is compared with the linear configuration by numerical simulations and phantom MR imaging. In-vivo study of healthy volunteer at 1.5T scanner was performed showing high potential of using such coil in clinical MR mammography.
The developed coil consists of two pairs of Helmholtz resonators forming a bilateral configuration (Fig. 1a). One pair of Helmholtz resonators (Y resonators) has mostly vertical magnetic field component, the other one (X resonators) – horizontal component. Each resonator is basically inductively coupled to the neighboring one. It was shown earlier that compensation of mutual inductance between volume resonators leads to an increase in transmit efficiency of wireless breast coil [5]. Therefore, in the proposed wireless coil decoupling elements were used to compensate the mutual inductance (Fig.1a). In order to avoid the manual reference voltage calibration and ensure standard clinical workflow, passive detuning circuits were used (5 in Fig.1a). The equivalent circuit of the wireless coil is shown in Fig.1b.
Numerical simulations are performed in CST Microwave Studio 2022. The coil was simulated together with transceiver birdcage coil (Fig.2a) and a homogeneous phantom (ε = 70, σ = 0.19 S/m for the breast, and ε = 78, σ = 0.45 S/m for the body). Both linear and quadrature configurations of wireless coil were simulated.
MRI experiments were performed on a 1.5 T Siemens MAGNETOM Espree. In order to demonstrate the increase in SNR due to quadrature configuration, experiments with a homogenous phantom were carried out for two setups: bilateral linear coil with two Y resonators (Fig.3a) and for the bilateral quadrature coil (Fig.3b). The flip angle (FA) maps were acquired by (GRE) acquisitions (TR/TE=2000/4.76 ms, FOV=323 × 323 mm2, thickness=3 mm, matrix=128 × 128, FA=45◦/90◦) using a double angle method [6]. The phantom images were obtained with same pulse sequence for FA=90◦. For experimental studies the Rx-only decoupled quadrature wireless coil was assembled shown in Fig.3b. In-vivo study setup included specially designed ergonomic case together with the coil shown in Fig.4c. Invivo images were acquired using the Rx-only decoupled quadrature wireless coil and a GRE sequence: FA=12◦, TR/TE=10.5/2.38 ms, FOV=361 × 374 mm2, matrix=352 × 340, and thickness=1 mm. The simulated B1+/ - maps for linear and quadrature configurations are shown in Fig.2b. The mean |B1- | value in ROI for linear Rx-only coil is 1.3 uT and 2.43 uT for quadrature coil. In transmit mode the mean |B1+| value is 0.31 uT for linear coil, and 0.32 uT for the quadrature. The obtained experimental FA maps are shown in Fig.3c for linear Rx-only coil and for quadrature Rx-only coil (Fig.3c). The phantom images are presented in Fig.3 for linear coil(a) and for quadrature coil(b). The SNR value of linear coil is 552, and 912 for the quadrature coil. The obtained in-vivo image is shown in Fig.4d. Due to the obtained numerical and experimental results the wireless coil works effectively as an Rx-only coil thanks to the passive detuning circuits. Moreover using the quadrature configuration allows to improve SNR in 1.7 times compared to linear coil. This study demonstrates the first Rx-only quadrature wireless coil for bilateral breast imaging at 1.5T. The obtained results demonstrate the potential of using such a coil for MR mammography at platforms of different vendors.
Pavel TIKHONOV (Saint-Petersburg, Russia), Alexander FEDOTOV, Georgiy SOLOMAKHA, Anna HURSHKAINEN
14:14 - 14:16
#46382 - PG176 Stretchable and flexible RF coils for extremity imaging in a portable, low-field MRI system.
PG176 Stretchable and flexible RF coils for extremity imaging in a portable, low-field MRI system.
Magnetic Resonance Imaging (MRI) at low magnetic field strengths offers significant advantages such as reduced equipment costs and increased portability, which helps improve access to MRI services. However, these systems often produce images of lower quality, mainly due to reduced signal-to-noise ratio (SNR), longer scan times, and decreased resolution [1-3]. Among the components that could significantly influence image quality is the radiofrequency (RF) coil [4]. This study explores the development and evaluation of flexible, stretchable RF coils tailored for a 72 mT MRI system [1], specifically designed to conform the wrist. The aim is to enhance both coil sensitivity and operational efficiency [5,6].
We constructed six solenoidal RF coils (Figures 1a-1f), each 12 cm long with a diameter of 7 cm, using Litz wire to provide flexibility and stretchability [5,6]. Four of the coils had ten turns made with different Litz wires, while the other two were designed with the most flexible wires maximizing the number of turns, taking advantage of the nearly negligible proximity effect at approximately 3 MHz [4,6]. To build the coils, we placed a 3D-printed mold with a wavy surface (1 cm peak-to-peak) over a sock containing a 3D-printed inner piece (Figure 2). The Litz wire was manually stitched to the sock at defined positions to maintain its stretchable structure.
To evaluate performance, we measured the quality factor (Q) of the coils both in the testbench and inside a 72 mT portable MRI scanner [1]. Inside the scanner, Q was measured under unloaded and loaded conditions using different phantom volumes: 10 mL, 125 mL, and 500 mL. We compared these results to those of a standard solenoid coil used in the same MRI system (Figure 1g), which has 44 turns and measures 14.7 cm in length by 14.9 cm in diameter. Additionally, we calculated the coil efficiencies using a B1 calibration protocol, where the applied power (P) is 1.17 W, the gyromagnetic ratio is 42.56 MHz/T [7], and the excitation time corresponds to a 180º flip angle.
Using these coils, we scanned the 500 mL phantom with identical sequences across all coils, adjusting only the RF pulse duration to ensure the same flip angle. In addition, we performed in vivo imaging using a RARE sequence on both the most efficient stretchable coil (Figure 1d) and the reference solenoid. The acquisition parameters were kept constant at 1.5×0.7×11.7 mm^3 resolution and a 30 kHz bandwidth. Among the six stretchable prototypes, the coil shown in Figure 1d achieved the highest unloaded quality factor (Q = 161), while the standard coil had a slightly higher value (Q = 189). However, when loaded with the 500 mL phantom, both coils exhibited a similar Q of approximately 136. The stretchable coil from Figure 1d also demonstrated the highest efficiency at 271 μT/√W, outperforming the standard coil, which reached 120 μT/√W. All stretchable designs either matched or exceeded the efficiency of the reference solenoid, with values of 135, 120, 135, 164, and 181 μT/√W for the coils in Figures 1a, b, c, e, and f, respectively.
Phantom imaging results are shown in Figure 3a, using the most efficient stretchable coil. Figures 3b and 3c illustrate the signal intensity profiles along the longitudinal and transverse directions of the phantom, captured with each of the seven coils. The stretchable coil from Figure 1d consistently produced the strongest signal. Additionally, in vivo scans using a volunteer (Figures 4a, 4b) showed visibly enhanced image quality when using the stretchable coil (Figure 4d) compared to the reference coil (Figure 4c), despite both scans lasting 10 minutes and having the same spatial resolution. The improved efficiency of the stretchable coils is largely attributed to their ability to closely conform to the anatomy being imaged, which increases both the filling factor and overall sensitivity. This proximity to the target region results in better coil performance compared to the conventional solenoid. It should also be considered that the shorter length of the stretchable coils may play a role in this enhanced performance. Imaging experiments with phantoms and in vivo confirm that the proposed coil designs can generate stronger signals, suggesting a clear benefit for low-field MRI applications. This study confirms that flexible and stretchable RF coils can significantly improve image quality in low-field MRI scans of the extremities, potentially aiding in more accurate diagnosis of injuries. Despite these advantages, some limitations remain, such as sensitivity to motion during scans and deformation of the coil geometry, which can affect the quality factor. Future work will address these issues by exploring auto-tuning mechanisms and using higher grade materials, such as polytetrafluoroethylene (PTFE) thread for stitching and new base materials to enhance the robustness and mechanical stability of the coil.
Jesús CONEJERO (Valencia, Spain), Teresa GUALLART-NAVAL, Pablo GARCÍA-CRISTÓBAL, Rubén BOSCH, Eduardo PALLÁS, Lucas SWISTUNOW, José Miguel ALGARÍN, Joseba ALONSO
14:16 - 14:18
#46969 - PG177 A new 9-channel ¹H, 3-channel ³¹P calf coil for interleaved multi-nuclear studies of skeletal muscle at 7 T.
PG177 A new 9-channel ¹H, 3-channel ³¹P calf coil for interleaved multi-nuclear studies of skeletal muscle at 7 T.
Magnetic resonance spectroscopy (MRS) is an established tool for dynamic exercise studies of muscle metabolism. Combining ³¹P and ¹H MRS enables a more comprehensive view of both oxidative [1] and glycolytic [2] metabolic regimes but requires dedicated RF hardware with high SNR for both nuclei and a strong and homogeneous transmit field. The coil design presented here combines three transceiver dipoles, to provide good ¹H excitation at 7 T, a three-element ³¹P transceiver array and a separate six-element ¹H receive-only loop array, to maximize ¹H sensitivity. Previously [3], we implemented and tested the ¹H part of this coil, demonstrating 3.8× and 1.3× higher ¹H SNR, respectively, on phantoms than our previous custom ³¹P/¹H calf coil [4] and a 28-channel ¹H-only reference coil (QED Knee coil, Siemens, Erlangen, Germany). Here we add the three-element ³¹P transceiver array and evaluate the finished coil’s ¹H and ³¹P performance on phantoms and demonstrate its applicability in vivo.
The newly-developed three-layer coil setup is shown in Fig. 1, along with a picture of the coil in its housing. In addition to the geometric decoupling achieved by the relative element positioning, LC and LCC traps were necessary to limit coupling between the ¹H and ³¹P elements. To evaluate the coil’s ¹H performance, flip-angle maps, GRE images (TR = 9.2 ms, TE = 4.24 ms) and noise-only scans were acquired with a homogeneous phantom using a 7 T MRI (Magnetom, Siemens Healthineers, Erlangen, Germany). SNR maps were calculated using the pseudo-multiple-replica-method [5]. The coil’s ³¹P SNR was evaluated on a phantom with 100 mmol/l K₂HPO₄ by acquiring localized spectra a using a semi-LASER sequence [6] (TR = 5 s, TE = 60 ms, 2500 Hz spectral width). The 50 x 25 x 50 mm³ voxel was positioned at a 45° angle, in a position mimicking the gastrocnemius medialis of the left leg. SNR was calculated as maximum signal over the standard deviation of 700 points of a noise-only region [7] and averaged over 100 scans. The results were compared to the previous custom ³¹P/¹H calf coil.
To demonstrate the coil’s performance in vivo, a localized ¹H spectrum was acquired in resting gastrocnemius medialis of a healthy subject using STEAM [8] (TR = 3 s, TE = 5.68 ms, 32 acquisitions, VAPOR water suppression [9]). Additionally, phosphocreatine (PCr) and creatine CH₂ (Cr2) time courses were acquired with interleaved ¹H/³¹P MRS during a single rest-exercise-recovery protocol. This was performed using a DRESS ³¹P sequence [10] (acquisition delay = 6.7 ms) with 15 mm slab thickness and ¹H semi-LASER [6] (TE = 52 ms) with 15 × 15 × 25 mm³ voxel size. The subject performed two plantar flexion pushes between each acquisition (TR = 6 s) on an MR-compatible ergometer, the exercise was sustained for 5 minutes. The acquired spectra were phased per channel and combined with weighting by SNR, using an in-house python script. Spectra were quantified with AMARES [10] in jMRUI, fitting lipids, creatine CH₃, TMA and Cr2 at 3.95 ppm (as doublet) in the ¹H spectra, and PCr and Pi in the ³¹P spectra. ¹H B₁⁺ and SNR maps are shown in Fig. 2. For the depicted ROI, the new coil provides 2.8× higher ¹H SNR than the reference, as well as a stronger and more homogeneous transmit field. In the ³¹P phantom measurements, SNR was 471 ± 22 and 641 ± 26 for the new coil and the reference coil, respectively; thus the new coil's SNR was 27 % lower. An example in vivo ¹H STEAM spectrum from resting muscle and a time course of 31P DRESS spectra is shown in Fig. 3, along with the corresponding voxel and slab positions on a localizer image. Fig. 4 shows the time courses of PCr and Cr2 during rest, exercise and recovery from a single subject, with closely matching depletion kinetics, τ(PCr-d) = 37.0 ± 3.2 s, τ(Cr2-d) = 38.5 ± 2.8 s, and similar recovery time constants, τ(PCr-r) = 60 ± 3 s, τ(Cr2-r) = 75 ± 4 s. The phantom data show that the new coil achieves a large ¹H SNR gain compared to the reference, as well as improved transmission. Its ³¹P SNR is lower due to the increased sample distance to accommodate the ¹H receive array and increased volume coverage, primarily in the z-direction. The ¹H spectrum and Cr2 time course demonstrate the coil’s capability to acquire high-quality spectra from small single voxels in vivo. The results shown in Fig. 4 highlight the coil’s suitability for the primary target applications of dynamic, interleaved ¹H/³¹P MRS. Future work will include a quantitative investigation of the coil’s performance on multiple subjects. These results are very promising for future applications of the coil in advanced metabolic calf muscle studies. Combined with further sequence development, the strong ¹H performance will enable the quantification of metabolites with small concentrations and restricted visibility, such as lactate [2], generating new insights into metabolic processes at higher exercise intensities.
Veronika CAP (Vienna, Austria), Vasco Rafael Rocha DOS SANTOS, Kostiantyn REPNIN, Peter WOLF, Graham J KEMP, Roberta FRASS-KRIEGL, Martin MEYERSPEER
14:18 - 14:20
#46802 - PG178 An adaptive design of a hexagonally-structured artificial dielectric with a set of dipoles to tailor the local RF transmit field at 7T and 10.5T.
PG178 An adaptive design of a hexagonally-structured artificial dielectric with a set of dipoles to tailor the local RF transmit field at 7T and 10.5T.
One of the challenges at ultra-high field MRI is RF transmit field inhomogeneity that results in significant local signal drops and loss of diagnostic contrast. New metamaterial-based designs offer solutions to locally tailor the RF transmit field[1-5]. Our recent design[6] examined a new approach of an artificial dielectric (AD) comprising of a hexagonally-structured set of copper strips, that provides an efficient coverage and thus achieves high effective dielectric constant of the overall structure. Furthermore, our previous work[7,8] showed that an efficient metamaterial can be designed as a combination of a dielectric layer with a set of electric dipoles. In this study a design of AD and a combination of AD with a set of dipoles are studied. The length of the added dipoles was exploited to adaptively change the effective structure characteristics. Phantom simulations and measurements were performed at 7T and 10.5T MRI to demonstrate the achievable RF transmit increase with AD. In addition, in-vivo non-human primate scanning at 7T MRI was performed with the new AD to increase both the RF transmit efficiency and local signal-to-noise.
Artificial dielectric: The AD comprises of 2 shifted copper strips grids with a dielectric (relative permittivity, εr=2.6) in-between, that generates a network of capacitors. The hexagonally structured design was compared to a grid to demonstrate more efficient coverage and thus higher effective dielectric constant. Eigenmode solver was used to characterize the AD configuration and its resulting transverse-electric TE01 mode and its frequency. The εr of a dielectric with the same TE01 frequency as the AD was found. The dependence of the thickness of the middle dielectric layer on the effective permittivity of the AD was tested. For actual implementation a dual-shifted hexagonally-structured pattern was used to achieve effective relative permittivity of ~95. This setup included a dielectric layer of εr=2.6 with 400 microns thickness and dimensions of 128x174 mm2.
Adaptive design of AD and a set of dipoles: A combined design of AD and a set of dipoles with various dipole lengths was studied. The dipole length of 30, 36, 50, 80 and 156 mm was examined in a setup with final dimensions of 128x174 mm2, realized as an array of 5x7, 4x7, 3x7, 2x7 and 1x7 dipole units, respectively (see Fig.2). The resulting TE01 frequency and the increase in the RF transmit field were examined.
Experiments: GRE scans at 7T MRI with a setup comprising AD and a set of dipoles were performed to examine the effect of the dipole length on the local signal increase (including transmit and receive contribution). Phantom with brain mimicking tissue properties (εr=53, σ=0.3 S/m) was used. In-vivo non-human primate scanning was performed to improve the local signal-to-noise and the RF transmit efficiency at 7T MRI. Both experiments were performed with a knee coil.
For our initial 10.5T work we utilized the 16Tx/80Rx head coil[9] and 16-rung shielded birdcage tune-up service coil (QED, Mayfield Village, Ohio, USA) available at all Siemens UHF sites[10]. Results with the head coil are shown, including B1 maps based on AFI[11]. Fig. 1A compares hexagonal and grid patterns of AD, demonstrating twice higher effective relative permittivity achieved with the hexagonal pattern. Fig. 1B shows that the same hexagonal structure can reach relative permittivity of >300 with dielectric layer of 100 microns (easily implemented with flexible PCB). Fig.2 shows eigenmode solver results for the combined AD and a set of dipoles. Shorter dipole length provides higher resonant frequency (and lower equivalent relative permittivity for the same dimensions dielectric with 7 mm thickness) and longer dipole length achieves lower resonant frequency (and higher equivalent relative permittivity). Fig. 3A shows the measured signal increase with a combined design of AD and a set of dipoles at 7T MRI, reaching an increase of 1.25-fold to 3.7-fold with the different dipole arrays. Since an array of the 156 mm dipoles (1x7 array) created a resonant structure at 298 MHz, it reached the maximal signal increase. Fig.3B shows that the same AD structure as in Fig.3A achieved maximal signal increase of 1.5-fold at 10.5T with the head coil setup.
Finally, in-vivo scanning of the non-human primate showed local signal increase of 1.3-fold (Fig.4) and 10% reduction in reference amplitude. A new hexagonally-structured AD provides compact and thin setup that can be easily incorporated in a realistic MRI environment. The AD setup was tested at 7T and 10.5T demonstrating a local increase of the RF transmit field. A combined design of AD with a set of dipoles can offer additional control over the resulting RF transmit increase, which can be used to tailor the field depending on the patient dimensions and different applications. Next steps will include careful evaluation of the SAR implications and the potential benefits at 10.5T.
Santosh K MAURYA, Alexander BRATCH, Edna FURMAN-HARAN, Evgeniya KORNILOV, Noam HAREL, Gregor ADRIANY, Rita SCHMIDT (Rehovot, Israel)
14:20 - 14:22
#47323 - PG179 Transceiver Coaxial-End Dipole Array for whole Brain and C-spine MRI at 9.4T.
PG179 Transceiver Coaxial-End Dipole Array for whole Brain and C-spine MRI at 9.4T.
Simultaneous brain and C-spine MRI at ultra-high fields (UHF, >7T) could provide more in-sights into the operation of the central nervous system. However, specially designed dedicated transmit (Tx) and receive (Rx) arrays are necessary to provide coverage over the whole region of interest. For combined head-neck imaging at 7T, recently, several loop-based transmit-only/receive-only (ToRo) (1) and transceiver (TxRx) (2,3) arrays have been proposed. Using loops for arrays requires many capacitors to be distributed along the loop to reduce SAR and sensitivity to the array loading. In this work, we designed a dual-row TxRx 16-channel coaxial-end dipole array to overcome these two issues. Our numerical and experimental results show that the proposed array can provide entire brain and C-spine coverage.
First, an anatomically shaped holder for a tight-fit array was designed (Fig.1A) using Siemens NX. Then, 16 coaxial-end dipoles (4) were distributed in two rows to cover the region of inter-est (i.e., brain and C-spine). For all numerical simulations, CST 2021 were used. Dipole ele-ments were constructed from a 1.6 mm diameter copper wire with 20 mm pieces of 50 Ohm coaxial cable with a 25 nH inductor in the end of coaxial. The ends of the dipoles were folded similarly to the dipoles in work (5) to reduce the dipoles' matching sensitivity to load variation. Duke and Ella voxel models were used for simulation. All array elements were matched to 50 Ohm using a CST Studio schematic module with an network of two series inductors and a par-allel capacitor. B1+, pSAR10g, COV, and SAR-efficiency were calculated for a set of CP-modes with different phase shifts between the array rows to define the best excitation mode.
The array housing was 3D printed from polycarbonate material. Milled polycarbonate plates were used to create the coil housing (Fig.1C,D). 16 BNC connectors were placed in the bottom upper flange of the coil to connect it with the TxRx interface (Fig.1B). Dipole array elements were made using 1.6 mm thick copper wire. Coaxial ends were made using non-magnetic RG-405 cable. The coaxial ends and the matching networks used self-made inductors from 1.2 mm diameter copper wire. The matching network in the experiments was similar to the one used in the simulations. Two floating ground cable traps (6) were placed between the dipole input and the BNC connector to prevent the cable effect.
A volunteer study was conducted after the array was constructed and tuned on-bench. All data were acquired using Siemens 9.4T full body scanner. In-vivo T1-weighted images were ac-quired using a 1 mm MPRAGE (7) (TR/TI=3.36 s/1.34 s, GRAPPA 2x2, matrix 364x242x192, adiabatic inversion pulse (HS4), FA=9°, BW=312 Hz/pixel). B1+ mapping was performed using the pre-saturated TurboFLASH (8) (TR/TE=10s/2.1ms, GRAPPA 2, FOV=380mmx262 mm, 28 sagittal slices of 3 mm thickness, base matrix size 64, in-plane resolution 6mmx6 mm, satura-tion FA=90°, excitation FA=8°, bandwidth=485 Hz/pixel). Based on the known reference volt-age, the acquired flip angle maps were converted into maps of B1+ per input power. The geo-metric distortion correction implemented by the vendor was applied to all reconstructed images to compensate for gradient non-linearity over the large field of view. The fully measured S-matrix of the array loaded with volunteer head and chest at 400 MHz is presented in Fig.1E. Fig.2 shows numerically simulated B1+ for the Duke (A) and Ella (B) and different phase shifts between the array rows. The presented distribution shows that the pro-posed array can provide a sufficiently homogeneous excitation within the ROI. Figure 3 shows the effect of different phase shifts between the array rows on the mean B1+ over the ROI, COV, pSAR10g, and SAR efficiency, based on simulations. Measured B1+ and MPRAGE images are presented in Fig.4. For simplicity, measurements were done for a 0° phase shift b/wthe rows. From the presented results, we can see that an increase in phase shift to 90° between the rows increases the mean B1+ in the ROI. However, this leads to an increase of the COV, i.e., a drop in field homogeneity over the ROI. According to the simulation results, the optimal phase shift is roughly 30°. It provides minimal COV and pSAR10g compared to all other phase shifts. Figure 4 shows that the proposed array can provide excitation of the whole brain and down to the C7 region. However, there are certain image artifacts due to motion, physiology, and B0 inhomogeneity in the neck/throat region. Advanced B0 shimming (9) and dynamic parallel transmission methods (10,11), such as kT points (12), could be used to improve excitation homogeneity in the future. We designed, constructed, and evaluated a 16-channel coaxial-end transceiver array for combined brain and C-spine imaging at 9.4T numerically and experimentally. We obtained images of the whole brain and C-spine in the experimental study down to the C7 region.
Georgiy SOLOMAKHA (Tübingen, Germany), Felix GLANG, Markus MAY, Joshi WALZOG, Dario BOSCH, Klaus SCHEFFLER, Harald QUICK, Nikolai AVDIEVICH
14:22 - 14:24
#46096 - PG180 Optimization of SNR at Both Frequencies for an UHF Double-Tuned Array: 7T 32-Element Transceiver 31P/1H Loop/Dipole Human Head Array.
PG180 Optimization of SNR at Both Frequencies for an UHF Double-Tuned Array: 7T 32-Element Transceiver 31P/1H Loop/Dipole Human Head Array.
X-nuclei (13C,31P etc) MRI and MRS provide valuable information for biomedical research and can benefit from the SNR increase at ultra-high field (UHF, 7T and above). UHF, however, brings the necessity of local transmission (Tx) and reception (Rx) capabilities within the structure of the same double-tuned (DT) RF coil. Hence, in the case of the double-layer Tx-only/Rx-only (ToRo) design (1), the DT coil must include four layers (2 Tx and 2 Rx) interacting with each other. In practice, such a complicated design has rarely been used. Use of transceiver (TxRx) arrays greatly simplifies the DT array by decreasing the number of layers to two (2-5). Still, to minimize interaction, often 1H-layer is moved away from the sample (2,3). This decreases the 1H SNR (2). Having good performance at both X and 1H frequencies is very important. Recently, we demonstrated that by placing both the 1H and 31P loop arrays into the same tight-fitting layer preserves the 1H Tx-efficiency and central SNR (5). However, we had to reduce the total number of loops to 20. Further improvement in SNR requires more Rx elements, which is very difficult to realize using common loops due to the design complexity. At UHF, the array design can be greatly simplified by using 1H dipoles (6). Combining 8 TxRx loops (X-nuclei) with 8 TxRx dipoles (1H) for human head imaging has been reported at 9.4T (7) and 7T (8).
In this work, we developed a novel UHF densely-populated tight-fitting loop/dipole DT array in which we increased the number of TxRx elements, all placed in one layer, to 32.
The developed 31P/1H 7T human head array consisted of two rows (2x8) of 16 31P loops (Figs.1A,1C) and 2x8 array of 1H dipoles (Figs.1B,1D). The entire coil measured 195 mm (left-right), 225 mm (posterior-anterior), and 210 mm (superior-inferior). Adjacent loops located in the same rows and different rows were decoupled by transformers and overlapping (Fig.1A), respectively. Coaxial-end dipole (9) measured 130 mm (without coaxial ends) in length. To minimize sensitivity of the resonance frequency to loading (10), the coaxial ends were moved away from the sample (Fig.1B). All reported data were acquired on a Siemens Magnetom 7T human MRI. The developed coil was compared to a commercial array coil (Rapid Biomedical) consisting of a multi-channel 31P Rx-array and quadrature TxRx 1H volume coil.
Electromagnetic simulations were performed using CST Studio Suite 2024 (Dassault Systèmes) and the time-domain solver based on the finite integration technique. We used a human voxel model (Duke, ITIS Foundation). We evaluated Tx-efficiency ( /√P) and SAR-efficiency (/√pSAR10g), where pSAR10g is peak SAR averaged over 10g of tissue (SCT Legacy averaging). As demonstrated previously (11), the Tx-performance of the 2x8 array is improved by a phase shift between the rows, which was also evaluated. In simulations, both arrays were evaluated separately (Figs.1A,1B). Fig.2 shows examples of simulated B1+ maps and corresponding quantitative data for the Tx-performance. Introduction of a phase shift between the rows improves the Tx-performance at both frequencies. After constructing, we evaluated coupling between all the array elements (Figs.1E,1F) with strongest coupling measured between adjacent elements. Worst between adjacent 31P loops measured -15.6 dB (same row) and -14.1 dB (different rows). Worst between adjacent 1H dipoles measured -14.9 dB (same row) and -14.3 dB (different rows). Isolation between 31P loops and 1H dipoles at both frequencies measured -30 dB or better. Fig.3 shows GRE images and corresponding 1H SNR maps obtained on a healthy volunteer using the developed and commercial array coils. The developed array provided substantially (~5 times) higher SNR peripherally and ~1.2 times higher SNR near the center. Fig.4 demonstrates the X-nucleus performance for a 31P MRSI measurement on the same volunteer. Despite the smaller number of Rx elements, the developed array provides similar 31P SNR to the commercial coil. At the same time, the new array greatly improves SNR at 1H frequency. Based on our experience, 2x8 transmit loop arrays can be well decoupled only at relatively high frequency, e.g. ~120 MHz and above, due to strong coupling between non-adjacent loop elements. This implies that at 7T, only 31P (possibly 7Li) arrays can be built using such a design. In addition, at 300 MHz, the performance of relatively short dipoles is sub-optimal in comparison to loops. At higher fields, e.g. >9.4T, 2x8 dipole arrays perform better (13). Also, more nuclei will exceed the 120 MHz limit. We demonstrated feasibility of constructing of a densely-populated UHF human head DT array with 32 TxRx elements all placed in one layer. First results show comparable 31P SNR at the 31P-frequency and a substantial improvement of 1H SNR compared to the existing state-of-the art commercial coil. The developed design can be even more beneficial at higher magnetic fields.
Nikolai AVDIEVICH, Georgiy SOLOMAKHA (Tübingen, Germany), Felix GLANG, Tanja PLATT, Stephan ORZADA, Dario BOSCH, Mark LADD, Andreas KORZOWSKI, Klaus SCHEFLER
14:24 - 14:26
#45527 - PG181 Numerical Comparison of TxRx arrays for combined Brain and C-spine MR Imaging at 7T.
PG181 Numerical Comparison of TxRx arrays for combined Brain and C-spine MR Imaging at 7T.
Combined brain and C-spine imaging at 7T can provide more detailed insights into various pathologies of this body region by increasing SNR compared to clinical 3T MRI(1). However, RF excitation of this region requires a local Tx-array providing a homogeneous excitation over the entire ROI (~350 mm longitudinally), which is a very difficult task at ultra-high fields (UHF, B0≥7T) because of the strong inhomogeneity of Tx RF magnetic field, B1+. The B1+ inhomogeneity can be mitigated using multi-element and multi-row Tx arrays in conjunction with 3D RF shimming(2,3). Recently, several works attempted to extend the Tx-coverage over the brain and C-spine using different array designs consisting of striplines(4) and loops(5). Dipoles array elements were introduced about 10 years ago(6), are a alternative to loops and striplines with simplified design. Following the work(7), we developed a coaxial dipole array for brain imaging at 9.4T(8). The coaxial dipole antennas improved the current distribution along the dipole, decreased local SAR, and minimized the frequency shift due to variation in head sizes compared to the straight dipoles. Coaxial dipole design could be even further simplified using coaxial-end dipole design(9). In this work, we numerically compared three transceiver (TxRx) arrays for combined brain and C-spine MRI at 7T regarding their Tx-efficiency, SAR, excitation homogeneity, and SNR.
We compared three arrays: 16 coaxial-end dipoles, 8 striplines(4), and 8 loops(5). The coaxial-end dipole array consisted of elements distributed in two rows on surface of a holder. All dipole elements were made using 1.2 mm wire and short coaxial cables. The longitudinal length of the dipoles in the upper row was 225 mm, and in the bottom row, 145 mm. To reduce load sensitivity, the ends of the dipoles were folded and moved away from the load. The length of the folded part was 25 mm, and the length of the coaxial ends was 20 mm. Eight stripline elements were arranged in a 6+2 configuration, with six elements on the top row around the head and two in the C-spine region. Stripline element and array geometries were identical to those described in the work(4). The loop array was arranged in a similar 6+2 configuration. The number of capacitors, element size, and their position were similar to those described in (5).
All numerical simulations were performed using CST 2021. Arrays were loaded with the Duke voxel model. All array elements were tuned and matched to 297.2 MHz. Isometric views of all three arrays are presented in Fig.1. All elements were matched to the -30 dB level. All arrays were evaluated using the CP-mode excitation. The coefficient of variation (COV) was used as a metric for B1+ field homogeneity. For this, magnetic field distributions were exported to MATLAB 2024, where the mean and standard deviation of B1+ were computed over a region that includes the whole brain and C-spine down to C7 (“ROI”), and for the C1-C7 region only (“SC”). SNR calculations were made using the sum-of-squares method(10). Simulated B1+ in the central sagittal slice of the Duke are presented in Fig.2. Dipoles improved B1+ performance compared to the stripline and loop arrays. The over ROI was 1.28 times higher than for striplines and 1.05 times higher than for loops. The over “SC” was 2.04 times higher than for striplines and 1.74 times higher than for loops. The pSAR10g values were similar for the dipole and loop arrays (0.339 and 0.325 W/kg), while pSAR10g was ~4 times higher for the stripline array. SAR-eff., calculated over the whole ROI for loops and dipole, was very similar (0.674 and 0.646 μT/√W/kg), while for the stripline array, it was ~2 times lower (0.267 μT/√W/kg). All three arrays showed very similar performance in terms of COV over the whole ROI, where the striplines showed the lowest variation (COV = 0.288) and the loops configuration showed the highest variation (COV = 0.349). The numerically calculated SNR is presented in Fig.3. The dipole array showed a significant increase in mean SNR over the whole ROI compared to the stripline (2.64 times higher) and loop array (1.91 times higher). The 16-channel folded-end dipole array outperforms other considered configurations of TxRx arrays for combined brain and C-spine imaging regarding Tx and SAR efficiency. In addition, the dipole array produces a significantly higher B1+ field in the spinal cord than both 6+2 arrays. Therefore, using a 16-channel configuration is preferable to a 6+2 one. Finally, the SNR of the dipole array was also higher. Since the majority of modern 7T systems have only 8 channels, to drive a 16-channel array, power splitters need to be used. In the next step, we plan to compare the proposed array's Tx performance with the 16Tx64Rx loop array(11). Three TxRx array designs for combined brain and C-spine MRI at 7 T. The 16-channel array showed improved performance both in Tx and Rx modes compared to 8-channel striplines and loop arrays.
Georgiy SOLOMAKHA (Tübingen, Germany), Markus MAY, Felix GLANG, Oliver KRAFF, Klaus SCHEFFLER, Nikolai AVDIEVICH, Harald QUICK
14:26 - 14:28
#47819 - PG182 Design and evaluation of a 16-channel Tx array for 14 T head imaging using simulations and H-field measurements.
PG182 Design and evaluation of a 16-channel Tx array for 14 T head imaging using simulations and H-field measurements.
In the MR community, increasing B0 field strength has been a key goal due to the expected gains in signal-to-noise ratio (SNR) and contrast-to-noise ratio [1]. The Dutch National 14 Tesla MRI Initiative (DYNAMIC) aims to establish the world’s first 14 T MRI system [2]. In this endeavour, both the main magnet and the remaining MRI components will be developed in parallel. However, RF coil validation will be challenging without a B0 field. Since electronic and surrounding losses are hard to compute, SAR simulations often assume experimental RF power normalization via B1+ mapping. Here, we propose an alternative approach using lossless RF antenna arrays that match the simulated geometry, decoupled cable management, controlled multi-transmit drive, and quantified H-field probes. This work presents the design and evaluation of a 16-channel Tx array for 14 T head imaging using simulations and absolute H-field measurements.
At 596 MHz, potential dielectric losses in construction media will be significant, particularly close to the antenna. Therefore, wire-wound antennas were constructed at least 1cm from the coil frame. S21 measurements were performed at a fixed distance between the antenna and the H-field probe with gradually increased distance between the antenna and the coil former to confirm lossless effects of the former at 1cm distance. A transmit array was constructed by placing sixteen of these spiralled dipoles on a 3D printed spacer Figure 1A). The antennas were placed in two rows of eight (causing two ground planes), staggered with ¼ antenna length overlap. The dipoles were fed at the center using a 50-Ω voltage source. Cables were routed via the ground planes to the radial extent of the coil housing and then routed to the top of the array. Each cable passed a tuned cable trap close to the antenna port, all aligned in the ground plane. No decoupling circuitry was added to the antenna array.
FDTD simulations were done in Sim4Life (Zurich MedTech, Switzerland) using the Duke model from the ITIS virtual family [3] (σ = 0.66 S/m, ε = 80) (Figure 1B). A convergence threshold of -50 dB was used. Phase shimming was applied to create constructive interference at the phantom center (Figure 1C). 10g-averaged SAR (SAR10g) maps were computed to assess SAR efficiency.
H-field measurements were done by placing a calibrated TBPS01 H-field probe (TekBox Digital Solutions, Vietnam) at the phantom center, which was filled with saline (Figure 2A). Two probe orientations were measured (0° and 90°). A 3 T MRI console (Philips, The Netherlands) generated a Tx signal. Eight up-mixers (Wavetronica, The Netherlands) converted 127.7 MHz to 596 MHz for transmission. The H-field probe output was down-mixed to 127.7 MHz for acquisition with preserved phase coherence (Figure 2C). Due to a current channel limit of 8, each dipole row was excited and validated separately. Simulations normalized to 16 × 1 W input yielded a B1+ hotspot of 0.84 μT at the phantom center, with a peak SAR10g of 0.28 W/kg and a SAR efficiency of 1.6 μT/√(W/kg).
Table 1 summarizes measured signal magnitudes and phases from the pickup probes at both orientations as measured during the transmit pulses. Individual antennas generated 0–1.95 nT. Without phase shimming, the top row (Tx1–Tx8) produced 3.91 and 1.56 nT; the bottom row (Tx9–Tx16) produced 0.78 and 1.30 nT. When phase-shimmed, the top row yielded 8.21 and 6.38 nT, and the bottom row 7.29 and 6.38 nT for the 0° and 90° probe orientations, respectively. The probe signal's resulting phase was zero when shimming was applied. Each antenna element had an input power of 0.69 mW during the measurements. The vector sum of the two orientations for both rows of the array results in a total field of 20.08 nT. Scaling it to 16 x 1W input power, this would yield 0.77 μT, close to the simulated B1+ of 0.84 μT. The agreement between summed sequential and simultaneously measured H-fields, and the zero phase in the shimmed field, confirms constructive interference at the phantom center and successful implementation of the multi-transmit system. The close match (92%) between measured and simulated B1+ magnitudes confirms low RF losses and successful coil design. Further refinement could correct for antenna mismatch, inter-element coupling, and contributions of B1-. This antenna array was derived from previously optimized designs for 596 MHz [4], promoting uniform excitation and low SAR. The correlation between simulation and experiment supports our method’s viability and paves the way for safe, reliable operation at 14 T. This work presents the design and evaluation of a 16-channel Tx array for 14 T head imaging using simulations and absolute H-field measurements without the presence of a B0 field. The array's ability to achieve focused B1+ fields through phase shimming that closely matches the RF simulations demonstrates low losses in the antenna setup and a robust multi-transmit bench setup.
Koen VAT, Esmé GALESLOOT, Alexander RAAIJMAKERS, Dennis KLOMP, Mark GOSSELINK (UTRECHT, Netherlands Antilles)
14:28 - 14:30
#47806 - PG183 Computational optimization of 3D-printed flexible MRI receive coils.
PG183 Computational optimization of 3D-printed flexible MRI receive coils.
Magnetic Resonance Imaging (MRI) is a widely used non-invasive technique in both clinical and research settings.[1], [2], [3], [4] Its effectiveness in demanding applications, such as functional MRI in animal models is often limited by low signal-to-noise ratio (SNR). The design of radio frequency receive phased array coils, commonly used to lower SNR, remains largely manual, and is constrained by fabrication complexity and poor anatomical conformity.[5], [6] While custom-built hardware offers potential improvements,[7] traditional methods struggle to accommodate subject-specific constraints that can be mitigated using novel manufacturing techniques such as 3D-printing.[8] To address these challenges, we present a surface-aware, parameterized modeling workflow that digitally optimizes the geometry of a two-channel array coil at 3T (Figure 1). Using Finite Element Method (FEM) simulations, the method iteratively adjusts the inter-coil distance to minimize coupling (S21 parameter) thereby improving the SNR.[5], [9] Applied to a cylindrical phantom, this approach yields measurable SNR improvements between initial and optimized designs (Figure 2). The optimized coils were 3D-printed in flexible resin and validated experimentally on a 3T MRI scanner, showing strong agreement between simulated and measured SNR maps. These results demonstrate a scalable path toward anatomically adaptive, high-performance RF coils for future (pre-)clinical MRI studies.
The studied design is a two-channel surface array coil composed of overlapping 40 mm loops conformally placed on a cylindrical phantom (100 mm length, 48 mm diameter). The coil geometry was parameterized by the ratio of the center-to-center distance between coils to their diameter (θ). A surrogate gradient-descent algorithm integrating MATLAB, Python, and COMSOL Multiphysics was used to minimize S21 and evaluate SNR across θ variations. Simulated SNR and SNR maps (Eq. 1) were computed using B1- (Eq. 2) and a noise correlation matrix derived from electric field distributions (Eq. 3). Coils were fabricated via stereolithography 3D-printing (Form 3BL, Formlabs) using Flexible 80A resin, forming hollow channels later filled with conductive liquid metal (GaInSn alloy). The phantom was printed in Clear V4 resin and filled with a gel composed of water, agar, benzisothiazolinone, and copper(II) sulfate. MRI experiments were conducted on a 3T MAGNETOM Prisma scanner (AS82 CPL gradient coil, 80 mT/m, 200 T/m/s) using a Turbo Spin Echo sequence at 1 mm³ isotropic resolution. Experimental SNR maps were calculated in Python using SciPy and NumPy by convolving a 3×3 kernel to estimate local noise from segmented image regions.
SNR = sqrt( B1-^T * R^(-1) * B1-^* ) (1)
B1- = (1/2) * (B1x - i * B1y) (2)
Rij = σ * ∫V [ Ei^* * Ej ] dv (3) A state-of-the-art design (θ = 0.75) and an optimized design (θ = 0.66) were successfully printed (Figure 2a). The algorithm converged in 20 iterations. Specifically the system improved from an initial state (θ = 0.75) of S21 = -4.82 dB and SNR = 1.37 x 10-5 to a better performance (θ = 0.66) with S21 = -18.8 dB and SNR = 1.75 x 10-5, showing an increase of 27.7% of the simulated SNR (Figure 2b top row). Measurements at the 3T scanner show a 25.3% improvement of the SNR for the same configuration (Figure 2b bottom row). A key challenge of this approach for more complex multi-parameter designs (e.g. 16-channels coils, fMRI head coils) is the high computational cost of detailed FEM simulations, especially with realistic loading and fine meshes. The surrogate-based gradient descent method offers an effective compromise, balancing accuracy with computational efficiency to achieve meaningful performance gains. This study presents a computational framework for optimizing conformal MRI surface coil arrays by minimizing inter-element coupling to enhance SNR. Combining parametric CAD, FEM simulations, and surrogate-based algorithmic optimization, we improved coil performance while ensuring manufacturability. The workflow was validated through 3T MRI and enabled fast, accurate fabrication using 3D printing. This multidisciplinary approach offers a scalable path toward personalized, high-performance hardware.
Quentin GOUDARD (Leuven, Belgium), Hanne VANDUFFEL, Cesar PARA-CABRERA, An VANDUFFEL, Dimitrios SAKELLARIOU, Uwe HIMMELREICH, Wim VANDUFFEL, Rob AMELOOT
14:30 - 14:32
#47642 - PG184 Multiple RF inputs and outputs for the open-source low-cost MaRCoS console.
PG184 Multiple RF inputs and outputs for the open-source low-cost MaRCoS console.
MaRCoS (Magnetic Resonance Control System) [1] is an open-source platform for controlling low-field MRI systems, based on the Red Pitaya SDRLab [2], which provides only two RF transmit/receive channels. This constraint restricts the implementation of essential multichannel techniques such as parallel imaging [3] and active noise cancellation [4,5].
To address this limitation in MaRCoS, we have developed a MIMO (Multiple Input Multiple Output) extension that enables synchronized operation of multiple SDRLab boards, with precise RF phase and amplitude coordination across all channels.
To synchronize the boards, we employ a shared system clocking where a master SDRLab board generates a reference clock distributed to slave SDRLab units. To coordinate the start of sequence execution, the master board also sends a digital trigger signal to the slave boards.
To validate the implementation of MIMO, we conducted two experiments. In the first test, we evaluated synchronization across multiple boards by running a script with a simple pulse sequence that generated an RF signal from transmit channels and routed it directly into receive channels. This test was repeated 100 times independently on two different hardware setups using up to three SDRLab boards (Fig. 1). Specifically, we connected the transmit channel of the master board to the receive channel of the first slave; the transmit channel of the first slave to the receive channel of the second slave; and the transmit channel of the second slave to the receive channel of the master board.
The second experiment assessed active noise cancellation on a mobile MRI scanner at the M-Tech lab. MIMO was integrated into the MRI4ALL [6] software to control transmission and reception. The setup used twisted-solenoid coils [7] for signal acquisition and five external sensing coils (5 cm, 10 turns) for environmental noise detection. Signals were pre-amplified and filtered (ZFL-500N+, 1–2 dB attenuators, BLP-2.5+ filters) before running a CPMG sequence. Multichannel data were processed using the EDITER algorithm to estimate and subtract environmental interference. Figure 2 summarizes the synchronization experiments. Figure 2.a shows the real and imaginary components of the signal acquired from the RX0 channels of the master (top panel), first slave (middle panel), and second slave (bottom panel) boards. Figure 2.b shows the amplitude (top panel) and phase (bottom panel) of the signal at the specific time instant highlighted in Fig. 2.a.
Figure 3 shows the results of the CPMG experiment used to evaluate active noise cancellation. The top panel displays the echo train acquired using a standard RF receive coil in the presence of environmental noise, showing clear signal degradation and fluctuation across echoes. The bottom panel presents the same dataset after applying EDITER, using the signals from the five additional sensing coils. Synchronization tests confirmed robust timing alignment, validating the hardware architecture and clock/trigger strategy. While the signal magnitude and phase varied between the boards, calibration can correct these differences. Active noise cancellation improved stability and signal recovery, demonstrating the potential of MIMO for multichannel interference suppression in low-field MRI. EDITER enhanced signal integrity by leveraging environmental noise estimations from the sensing coils.
We are now extending the noise cancellation to full image denoising, aiming to improve image quality in environments with high electromagnetic interference. In parallel, we are exploring parallel imaging techniques.
Ongoing work includes extending noise cancellation to image-level denoising and investigating parallel imaging methods.
To scale up, we are also developing a PCB for distributing the master clock to up to 32 Red Pitayas while preserving signal integrity. This will enable the deployment of high-channel-count receive arrays. The MIMO extension developed for MaRCoS enables synchronized multichannel reception using multiple SDRLab boards and the active noise cancellation experiments further demonstrated the practical utility of this setup. Existing solutions up to now were either proprietary or significantly more expensive [8].
José Miguel ALGARÍN, Aaron PURCHASE, Vlad NEGNEVITSKY, Luiz Guilherme DE CASTRO SANTOS (Valencia, Spain), Joseba ALONSO
14:32 - 14:34
#47657 - PG185 Scan2Go Project – an Assistive Brain Imaging Device to Enable Fast, Silent and Accessible Brain MRI for Dementia Patients.
PG185 Scan2Go Project – an Assistive Brain Imaging Device to Enable Fast, Silent and Accessible Brain MRI for Dementia Patients.
MRI is highly accessible due to its non-invasiveness. Yet, patient populations such as those with dementia or geriatrics can find MRI procedures difficult. Acoustic noise can cause miscommunication problems, induce panic and claustrophobia and disorientate dementia patients [1-4]. While the MRI patient table is also an obstacle, as climbing onto the table without assistance can be challenging for older age groups. Therefore, the current MRI system is unsustainable for use with such patient groups.
The Scan2Go Project aims to facilitate brain MRI to be more accessible for dementia and geriatric patients with a brain imaging device comprised of a movable chair, a silent gradient coil and an open Rx coil. Acoustic noise arises due to rapidly oscillating gradient coils during spatial encoding [5]. Using an ultrasonic gradient coil operating at 20kHz, above the human hearing limit, spatial encoding is effectively silent [6-10]. The patient table is replaced by a chair that can move the subject's head into the isocentre from a seated position, which does not require the subject to exert themselves. The open Rx coil eases the chair's and subject's mobility by providing greater space for the head.
This abstract presents the design of the brain imaging device as well as first scans of a phantom with the custom-made ultrasonic gradient coil and Rx coil.
The chair (Figure 1) was designed by INNO Metaal (Eindhoven, the Netherlands) and has a size of 1210x11270x1410mm^3 (WxLxH). The chair's height was chosen based on the 95th percentile of body sizes. An operator moves the chair manually with a tumble switch to a final position set with the console. The chair can also be detached in the case of evacuation. Futura Composites (Heerhugowaard, the Netherlands) designed the gradient and patient tube while the Rx coils were designed by Tesla Dynamic coils. Both coils are housed behind a custom patient tube, which guides the chair (Figure 1a) to the final position and leaves open space so the subject is less constrained (Figure 2). The gradient coil has an inner diameter of 554mm and length of 180mm; the Rx coil has a diameter of 388mm and length of 119mm. The gradient coil can be operated silently with a capacitor bank (Figure 3), making the coil resonant at 20 kHz in series with an NG500 gradient amplifier (Prodive, Eindhoven, the Netherlands). The gradient coil can achieve a maximum gradient strength and slew rate of 40mT/m and 5026 T/m/s, respectively. The device is designed for a 1.5T scanner and tuned to 64MHz.
The first images were acquired using a dual-echo multi-slice gradient-echo sequence with: FOV=320x320x219.6 mm3, in-plane voxel-size=4x4 mm2, slice-thickness = 4 mm, slice-gap = 0.4 mm, flip-angle = 12 degrees and TR/TE1/TE=9.3/2.9/6.6 ms. These were used to map the gradient field by supplying a 1 A DC-current to the ultrasonic gradient. Figure 4 shows the first images acquired with the Scan2Go-setup. Figure 4b shows the measured gradient field and the estimated efficiency (0.04 mT/m/A) of the ultrasonic gradient in the Scan2Go-setup. In summary, we presented a novel device that could be used seamlessly by a trained operator, allowing positioning of a patient into the scanner bore from a seated position. The design of the chair, including the tumble switch, ensured the operation of the chair matched that of a typical patient table, while safety considerations were also made to enable smooth evacuation as well as access to a nurse call button and headphones for communication with the operator (MCOM, Figure 1a).
The imaging experiments in this abstract demonstrated the first imaging with the Scan2Go setup and validated the gradient field of the ultrasonic gradient. The device can also be operated at 20kHz, above the human hearing limit, enabling fast and silent brain imaging. The Scan2Go Assistive Brain Imaging Device facilitates the positioning of less-abled patients to complete brain imaging protocols and enables fast and silent brain imaging. This device has the potential to make brain MRI more accessible for dementia patients as well as other physically disabled patient groups. The Scan2Go project also aims to evolve into a fully automated setup, which could reduce operational costs and streamline procedures for patients.
Michael MCGRORY (Utrecht, The Netherlands), Thomas ROOS, Edwin VERSTEEG, Mark GOSSELINK, Cezar ALBORAHAL, Thijs VAN HOOREN, Carel VAN LEEUWEN, Hans VAN DEN BERGE, Martin OOME, Wout SCHUTH, Martino BORGO, Jeroen SIERO, Dennis KLOMP
14:34 - 14:36
#46477 - PG186 Comparison of modular flexible and standard MR coils for thoracic outlet syndrome imaging at 3T.
PG186 Comparison of modular flexible and standard MR coils for thoracic outlet syndrome imaging at 3T.
Thoracic outlet syndrome (TOS) is characterized by upper limb pain caused by compression of neurovascular structures, typically in the interscalene space (ISS) and costoclavicular space (CCS). While TOS diagnosis is primarily based on clinical assessment, imaging is essential to localize and assess the severity of compression. Dynamic imaging, while not yet standard practice, has shown potential in a prior study using dynamic CT during functional maneuvers [1].
However, MRI is the preferred method for TOS imaging because of superior soft tissue contrast [2]. The addition of dynamic protocols could further enhance MRI’s diagnostic value but limits coil selection. Available standard coils offer good coverage but suboptimize signal-to-noise ratio (SNR) in potential compression zones due to their rigidity or semi-flexibility. Modular flexible coils, like the ModFlex coil [2], provide superior anatomical conformity and accommodate patient motion more effectively in dynamic MR exams.
This study compares the MR imaging performance of the 16-channel ModFlex coil and an 18-channel product coil in healthy volunteers at the two typical regions of neurovascular compression associated with TOS.
9 healthy volunteers (3 females, 6 males; age 30 ± 8 years) participated in one measurement session using two consecutive TOS MRI protocols: one with the 16-channel ModFlex coil [3] and another with the manufacturer’s 18-channel Body 18 coil on a MAGNETOM Prisma 3T (Siemens Healthineers, Erlangen, Germany). ModFlex coil positioning is shown in Fig. 1. Both protocols were conducted under ethical approval of the EDEN study (ClinicalTrials.gov NCT05218460, CPP SUD-EST IV, 26.07.21), with volunteers positioned supine with arms overhead within the opening of the MRI tunnel.
Each protocol included anatomical 2D T1w TSE sagittal sequences (0.48×0.48×3 mm³, FOV 142×160 mm²). 3D GRE (2.2 mm³) and noise-only scans were acquired for multi-channel SNR calculation using the pseudo-replica method [4].
Two bilateral regions of interest were manually approximated on SNR maps using 3D Slicer [5], based on identifiable anatomical structures: the ISS, following the estimated brachial plexus path between the anterior/middle scalene muscles and first rib; and the CCS, between the clavicle and first rib.
SNR values were extracted for each region and averaged within each ROI and across both sides. The resulting values were compared across coils using a Wilcoxon signed-rank test, with significance set at p<0.05. The SNR comparison results are shown in Fig. 2. In the CCS, the ModFlex coil demonstrated significantly higher SNR (p = 0.004). In the ISS, SNR was higher on average with ModFlex, but the difference was not statistically significant (p = 0.098).
Typical SNR maps of one volunteer acquisition are shown in Fig. 3, and representative anatomical images acquired with each coil are presented in Fig. 4. Anatomical details are clearly visible in ModFlex MRI whereas Body 18 coil images show noise influence in the region of interest. The higher SNR observed in the CCS likely results from the closer positioning of the coil to the region of interest. Variability in SNR measurements within the ISS may be influenced by anatomical differences among subjects and small variations in coil positioning; nevertheless, a trend toward improved SNR with the ModFlex coil was observed. A systematic study on the coil positioning could further improve its performance. Future work will include studies with larger cohorts and standardized protocols to validate these results. Using a modular flexible coil array at 3T MRI enables efficient assessment of thoracic outlet syndrome, with significant SNR improvements in the costoclavicular space and a trend of improved SNR in the interscalene space.
Catherine TRUONG (Nancy), Bouchra ASSABAH, Pedro TEIXEIRA, Audrey KIRSCH, Pierre-André VUISSOZ, Jacques FELBLINGER, Elmar LAISTLER, Lena NOHAVA, Karyna ISAIEVA
|
Salle 120 |
|
"Friday 10 October"
Added to your list of favorites
Deleted from your list of favorites
E24
14:00 - 15:30
ECRC
Spin Together: Aligning People and Practices in Research using Lab Handbooks
Keynote Speakers:
Beatrice LENA (Ph.D.) (Keynote Speaker, Leiden, The Netherlands), José MARQUES (PhD), Myrte STRIK (Postdoctoral Researcher) (Keynote Speaker, Amsterdam, The Netherlands), Benjamin TENDLER (Keynote Speaker, Oxford, United Kingdom)
|
Salle 76 |
|
"Friday 10 October"
Added to your list of favorites
Deleted from your list of favorites
G24
14:00 - 15:30
Poster 4
FT3 - Best evidence and best practice | FT2 - Cardiac and Vascular | FT2 - Tumours
14:00 - 15:30
#47795 - PG364 Infection Prevention and Control in Magnetic Resonance Imaging: Investigating Radiographer Knowledge, Attitudes, and Practices in Contrast Administration.
PG364 Infection Prevention and Control in Magnetic Resonance Imaging: Investigating Radiographer Knowledge, Attitudes, and Practices in Contrast Administration.
Magnetic Resonance Imaging (MRI) presents a unique infection prevention and control (IPC) challenge due to its unique environment, complex workflows and distinct requirements for MRI-safe equipment [1]. Despite the clinical importance of IPC [2], specific guidance tailored to MRI remains limited. This study investigates the knowledge, attitudes, and practices (KAP) of MRI radiographers related to IPC for intravenous (IV) contrast administration, including the influence of training, workplace policies, and perceived infection risks.
An online cross-sectional survey targeting MRI radiographers in Australia was launched in October 2024. The survey gathered demographic information and evaluated KAP domains using Likert-scale and multiple-choice items. Additional questions focused on the challenges of IPC adherence, the availability of contrast-related IPC policies, the time since the last formal IPC training, and primary sources of IPC knowledge. Survey responses were collated and analysed using Microsoft Excel. A total of 50 registered MRI radiographers completed the survey, 80% of whom were senior radiographers or MRI section leaders, and 76% had more than 10 years of experience in MRI. Study participants had high overall knowledge scores (93%) and positive attitudes (87%) toward IPC. However, only 66% always applied standard precautions when interacting with patients, with the remainder either frequently or occasionally applying standard precautions. Applying standard precautions was lower (52%) when utilising MRI equipment. Half (58%) of the participants had completed IPC training in the past year, while 46% and 14% either did not have or were unsure if they had an IPC team and IPC policies, respectively. Overall, in most of the participants’ workplaces, radiographers are responsible for setting the contrast injector, connecting the intravenous contrast to the patient, and cleaning the injector. However, only 26% of participants would clean the MRI equipment between patients, with 32% cleaning the room once a day. While institutional guidelines were commonly cited as the primary source of information, peer discussions were the most frequently reported source of knowledge regarding contrast injectors. Participants consistently identified MRI injectors, contrast tubing, and accessories as high-touch, high-risk equipment, with 70% believing their workplace recognised them as such. However, only 25% of participants would clean IV contrast-related equipment more than once per day. IPC adherence was reported to decline during high patient volumes, with respondents citing limited time to maintain aseptic technique during IV cannulation and contrast administration. These findings indicate that MRI radiographers have a strong foundational knowledge and a proactive attitude toward IPC during contrast procedures. However, inconsistent workplace recognition of high-risk surfaces and reliance on informal learning may undermine their practice. Time pressure and the challenge of maintaining aseptic technique during IV cannulation, particularly in busy or emergency settings, further affect the consistency of IPC. This study highlights a gap in MRI-specific IPC guidance, particularly regarding contrast administration. While radiographers demonstrate strong foundational knowledge and commitment to IPC, the lack of consistent, evidence-based protocols and modality-specific procedures undermines practice. Addressing these limitations requires a multifaceted approach, integrating formal training, MRI-appropriate IPC frameworks, and institutional support to promote consistent, safe, high-quality care. Radiographers are well-positioned to lead and inform these developments, and their active involvement in future research and policy design will be crucial to advancing IPC standards in MRI environments.
Frances GRAY (Sydney, Australia), Dania ABU AWAAD, Yobelli JIMENEZ, Suzanne HILL, Sarah LEWIS, Peter KENCH
14:00 - 15:30
#45630 - PG365 Student Perceptions of MRI Simulation in their Undergraduate Radiography Education.
PG365 Student Perceptions of MRI Simulation in their Undergraduate Radiography Education.
Simulation-based education enhances practical competencies ahead of clinical placements. In radiography, MRI simulation provides a safe, interactive environment for students to develop technical and decision-making skills. The addition of a scanning simulation tool was added in response to updated HCPC standards, which now require qualifying radiographers not just to assist but be fully capable of conducting an MRI examination. The aim of this study is to evaluate the initial experiences, perceptions, and acceptance of student radiographers in utilising an MRI simulator for their educational development.
A focus group with six final-year radiography students was conducted. Participants were self-selected volunteers who had completed an MRI simulation module. Discussions were transcribed and thematically analysed. Six overarching themes emerged: Curriculum structure, Technical Challenges, Guidance and Support, Confidence and Skill -Building, Learning process, clinical practice. Some noted improved engagement in clinical discussions and job interviews. However, technical and instructional gaps were identified, with students requesting deeper explanations of MRI parameters. Students also suggested earlier and more frequent integration to better align with theoretical learning and clinical placements. Students valued MRI simulation for developing competency and confidence but highlighted the need for earlier integration, improved usability, and stronger alignment with clinical practice. Findings support structured MRI simulation implementation to optimise learning outcomes. Future research should explore the long-term impact on competency retention and employer perceptions of simulation-trained graduates.
Ioele ELISABETH, Darren HUDSON (Exeter, United Kingdom)
14:00 - 15:30
#47493 - PG366 Assessment of 2D and 3D Cartilage Thickness Measurements in a Positioning Test–Retest Scenario.
PG366 Assessment of 2D and 3D Cartilage Thickness Measurements in a Positioning Test–Retest Scenario.
The cartilage thickness measurement from MRI images has become a vital part of clinical trials and longitudinal OA studies. With the availability of automatic segmentation tools, the assessment of cartilage has become quicker and more accessible [1]. However, thickness measurement is still challenging, as the cartilage tissue spans only 2–10 voxels in-plane, and the inclusion or exclusion of voxels at the boundary can significantly affect the results [2]. Additionally, the measured thickness can vary with slice positioning, making 3D thickness measurement a potentially better option to the 2D approach. While consistent patient positioning during follow-up visits improves overall precision, it may not always be possible due to factors such as pain. The objective of this experiment was to assess the repeatability of thickness measurements when the knee is rotated and to compare the 2D and 3D approaches.
The left knees of eight healthy volunteers (4 men, 4 women, mean age: 35.5 ± 10.2 years) were scanned on 3T Siemens PrismaFit (Siemens Healthineers AG, Forchheim, Germany). The 3D DESS (TE=5ms, TR=14.1ms, 160 slices, 0.6x0.6x0.6mm3, flip angle=25°, acquisition=5:58min) was used. Each volunteer's patella center was marked with a black line, and two additional lines spaced 1cm apart were drawn on each side (Figure 1). A dedicated 15-channel knee coil was positioned to align these lines with the scanner laser, producing five distinct knee positions: neutral, two medial rotations, and two lateral rotations. Knee Position Angles were measured using RadiAnt DICOM Viewer (Medixant, Poznań, Poland) (Figure 2).
Images were automatically segmented using MR ChondralHealth version 3.1 (Siemens Healthineers AG, Forchheim, Germany), with manual corrections applied when necessary. Nine femoral cartilage regions were assessed: medial (anterior/central/posterior), trochlear (lateral/central/medial), and lateral (anterior/central/posterior). Cartilage thickness was computed using a custom Python script. Cartilage voxels adjacent to the bone (proximal surface) were identified, and the real-world coordinates of voxel vertices shared between bone and cartilage at the bone-cartilage interface were calculated using the affine matrix. The distal surface voxel vertices were identified as vertices shared with the background (value 0) neighboring voxels. For each voxel along the distal surface, the nearest voxel along the proximal surface was found using a KDTree nearest-neighbor search (SciPy library [3]). Cartilage thickness was calculated as the mean of Euclidean distances between each distal surface voxel and its corresponding nearest neighbor on the proximal surface. This was performed either in 3D, or as 2D for each individual slice. The average thickness for each cartilage segment in 2D scenario was then obtained by averaging the thickness values across all slices within that segment. Slices with large discrepancies between the number of proximal and distal surface vertices, may result in overestimated cartilage thickness, therefore measurements exceeding 3 mm were excluded.
Finally, correlations between knee rotation angles and thickness were evaluated using Repeated Measures Correlation calculated with R library rmcorr [4]. Results are summarised in Table 1 and Table 2. Given the small number of probands and the large number of comparisons, the summary statistics presented in Table 1 should be interpreted as exploratory and primarily for orientation.
The relationship between rotation angle and cartilage thickness varies across femoral regions and differs between 3D and 2D measurements. Notably, the trochlea medial segment shows a strong negative correlation in 3D, which is not observed in 2D. This suggests that precise and consistent positioning is essential when using 3D thickness measurements, particularly in longitudinal follow-up studies. Overall, cartilage thickness appears to be influenced by knee rotation in most femoral regions, with the exception of the medial posterior femur, where no correlation was observed. This study demonstrates that femoral cartilage thickness is influenced by knee rotation, with region-specific patterns of correlation. The findings underscore the importance of consistent joint positioning in imaging protocols, especially for longitudinal assessments. Notably, the absence of correlation in the medial posterior region suggests localized variations in how rotation impacts cartilage morphology.
Veronika JANACOVA (Vienna, Austria), Pavol SZOMOLANYI, Diana SITARCIKOVA, Vladimir JURAS
14:00 - 15:30
#45479 - PG367 The diagnostic utility of neuromelanin-sensitive MRI in Parkinson’s disease: a systematic review.
PG367 The diagnostic utility of neuromelanin-sensitive MRI in Parkinson’s disease: a systematic review.
Parkinson’s disease (PD) diagnosis primarily depends on clinical criteria, but the need for objective biomarkers remains critical due to limitations in early and accurate detection. Neuromelanin-sensitive magnetic resonance imaging (NM-MRI) has emerged as a non-invasive imaging modality capable of visualising neuromelanin depletion within the substantia nigra pars compacta (SNpc), a hallmark of PD pathology. While initial studies suggest NM-MRI holds promise for enhancing diagnostic accuracy, considerable methodological variability presents a challenge to its integration into clinical practice. This systematic review focuses on evaluating NM-MRI’s diagnostic potential, examining technical variability, and considering its future role in PD diagnosis.
We systematically reviewed 29 diagnostic accuracy studies encompassing 1,207 patients with PD and 990 controls. Studies predominantly utilised 3T MRI scanners with T1-weighted fast spin-echo sequences, though considerable variation existed in protocols, image analysis techniques (manual, semi-automated, automated), and target anatomical regions within the SNpc. Diagnostic metrics- sensitivity, specificity, and area under the curve (AUC)- were extracted and rigorously analysed to determine NM-MRI's diagnostic performance. NM-MRI consistently exhibited high diagnostic sensitivity across studies, with values ranging from 60% to 100%; notably, approximately one-third of studies reported sensitivities above 90%. Specificity varied more significantly, from 66.7% to 100%, with over 85% of studies reporting specificity exceeding 80%. Robust AUC values (0.80–0.99) were frequently reported, particularly when employing volumetric analyses of SNpc. Despite these strong diagnostic metrics in identifying PD vs healthy controls and even atypical parkinsonian syndromes (APS), significant methodological heterogeneity—including variations in MRI acquisition parameters, segmentation techniques, and clinical populations studied—contributed to observed inconsistencies in performance. This systematic review evaluates NM-MRI's diagnostic utility for Parkinson’s Disease, highlighting strengths such as a high number of comparative studies and consistent use of validated diagnostic frameworks. However, methodological limitations like variability in imaging protocols and small-scale studies affect generalizability. Clinical implementation faces challenges due to the lack of standardized protocols; adopting T1-weighted fast spin-echo sequences and 3T MRI platforms is recommended. The SNpc region, especially its lateral part, is key for diagnosis, and combining analyses with the LC enhances accuracy. Workflow integration requires balancing scan duration and analysis methods, with automated tools showing promise. Standardization across technical parameters and analysis methods is critical for consistency. Although resource-intensive, NM-MRI could offer long-term cost savings through improved diagnostic accuracy. Future research should focus on achieving international protocol consensus, conducting large-scale validation studies, and integrating NM-MRI with AI and multimodal diagnostics for comprehensive PD management. NM-MRI demonstrates robust potential as a sensitive and reliable biomarker for PD diagnosis and differential diagnosis. Addressing methodological variability through protocol standardisation and rigorous multicenter validation is essential to optimise its diagnostic accuracy, reproducibility, and integration into routine clinical practice.
Rayo AKANDE (Basel, Switzerland)
14:00 - 15:30
#47901 - PG368 Localising unilateral lumbosacral radicular pain through Diffusion Tensor Imaging: an experimental study.
PG368 Localising unilateral lumbosacral radicular pain through Diffusion Tensor Imaging: an experimental study.
Unilateral lumbosacral radicular pain is common, but accurately diagnosing it remains difficult [1]. Conventional macroscopic anatomical MRI (T1- and T2-weighted sagittal and axial sequences) produce a notable number of false positives or false negatives, resulting in discrepancies between MRI findings and patient symptoms [2-7]. In contrast, microstructural MRI, such as Diffusion Tensor Imaging (DTI), can reveal pathological changes in the form of reduced fractional anisotropy (FA) in the affected lumbosacral nerve, even in the absence of overt nerve compression [6-12]. However, there is no consensus yet on the optimal DTI protocol for reliably identifying symptomatic nerve roots [13].
In this experimental study, we aim to identify the most reliable DTI acquisition strategy for the robust visualization and segmentation of the lumbosacral nerve roots (LNR) (L3–S1), with the ultimate goal of enabling quantification of the extent of unilateral damage associated with radicular pain. We scanned healthy controls using multiple spinal cord DTI protocols and evaluated how well each protocol preserved anatomical normality. Additionally, we examined the effect of two distinct DTI processing pipelines to assess the relative impact of pipeline’s strategies on tractography results.
Seven healthy volunteers were scanned on a 3T Siemens Prisma system at the GIGA In Vivo Imaging platform, GIGA Institute, University of Liège, Belgium. Three different DTI protocols were considered (Table 1). ZOOMit showed higher anatomical detail but longer scan time (~35 min), while Coronal was fastest (~7 min). Due to time constraints, only two protocols were applied per subject, with each tested in four volunteers (Table 2).
Two different DTI pipelines were examined to evaluate the impact of preprocessing on tractography: “SCT” and “our”. The former uses the MOCO algorithm [17], while the latter consists of denoising [19], correction for Gibbs ringing artifacts [20], correction of distortions due to susceptibility, head motion, and eddy currents [21]. “None” refers to a baseline case with no preprocessing. Tractography was performed using MRtrix3's tckgen algorithm [22]. We estimated FA using DTIFIT in FSL with weighted linear least squares [23]. The FA symmetry score was computed by multiplying the FA map and tract density map voxel-wise, then summing the weighted values separately for the left and right sides of the spinal cord (Figure 1). Figure 1 shows left–right FA symmetry for the L3 to S1 nerve roots. Results are grouped by DTI protocol (Standard axial, ZOOMit axial, Coronal) and preprocessing pipeline (none, SCT, our). Substantial differences are observed across both protocols and pipelines. The combination of ZOOMit and our pipeline provided on average the highest FA symmetries for L3, L4, and L5. However, the Coronal DTI protocol combined with the our pipeline provided the highest FA symmetries for S1.
Figure 2 shows representative tractography results of the lumbosacral nerve roots using the three examined DTI protocols. Notable differences in streamline density and anatomical coherence are observed across protocols, with ZOOMit axial DTI yielding the highest coverage but also more spurious tracts. This study shows that the choice of DTI acquisition protocol has a substantial impact on tractography outcomes and left–right FA symmetry. The visibility and reconstruction quality of specific lumbosacral nerves varied across protocols: the ZOOMit axial DTI provided the most consistent and detailed tractography for the L3 nerve, while both the ZOOMit and Standard axial protocols performed comparably well for L4 and L5. In contrast, the S1 nerve was reconstructed with similar reliability across all three protocols. Notably, the ZOOMit protocol exhibited increased sensitivity to spurious streamlines, emphasizing the need for careful parameter tuning to minimize false positives. Between the two preprocessing pipelines evaluated, our in-house method yielded slightly better performance than the SCT pipeline. While this experimental analysis was conducted on a limited number of subjects, it offers preliminary results toward the optimization of DTI-based imaging of the lumbosacral nerve roots. Future studies involving larger cohorts are warranted to validate these findings and to further refine protocol selection for clinical and research applications. In conclusion, our findings highlight that the choice of DTI acquisition protocol has a more significant impact on tractography and FA symmetry of the lumbosacral nerve roots than the choice of processing pipeline. ZOOMit provided the best anatomical detail for L3–L5, while all protocols performed similarly for S1. Careful tuning of tractography parameters and thoughtful pipeline selection remain essential.
Evgenios KORNAROPOULOS (Liège, Belgium), Pierre PESESSE, Mark VANDERTHOMMEN, Christophe DEMOULIN, Laurent LAMALLE, Christophe PHILLIPS, Mikhail ZUBKOV
14:00 - 15:30
#46765 - PG369 From Magnetic Gradients to Sound Waves: A Five-Year Journey of the ESMRMB Podcast.
PG369 From Magnetic Gradients to Sound Waves: A Five-Year Journey of the ESMRMB Podcast.
Effective science communication is increasingly recognized as a critical component of responsible research practice, fostering transparency, enhancing public trust, and supporting reproducibility and collaboration across disciplines. Prior studies have shown that open access dissemination and targeted communication strategies can improve research rigor, democratize knowledge, and accelerate cross-border collaboration. In this spirit, the ESMRMB Early Career Researchers Committee launched the ESMRMB Podcast in 2020 to create an accessible and sustainable platform for the scientific community. This ongoing endeavor is entirely developed, hosted, and managed by early career researchers, with the overarching goals of enhancing knowledge exchange, fostering collaboration, and advocating for effective science communication. Additionally, the podcast aims to promote diversity in skillsets, academic backgrounds, career paths, and perspectives within the MR field.
Here, we present a descriptive analysis of listener engagement, demographics, and global reach of the ESMRMB Podcast over a five-year period, highlighting the impact and value of this podcast for community-driven science communication.
A total of 16 episodes were recorded between October 2020 and April 2025 via Zoom with invited speakers, and edited using Apple iMovie. A total of 16 episodes were released, featuring content across a broad range of MR-relevant themes including MR technologies, clinical applications, career development, networking, grant writing, science communication, open access, international collaboration, and diversity, equity, and inclusion. The podcasts were distributed through major platforms such as Apple Podcasts and Spotify. Listener analytics were collected and evaluated over five years, including engagement trends, demographics, geographic distribution, device and platform usage. Listenership remained consistent, with peaks typically aligning with new episode releases (Figure 1). The majority of listeners were aged 23–44 (87.3%) and identified as male (62.7%), reflecting strong engagement from early-career professionals and researchers (Figure 2). The podcast reached a global audience, with top listener countries including the United States (23.6%), United Kingdom (15.1%), Germany (14%), the Netherlands (8.3%), and Japan (7.9%) (Figure 3). Multi-platform access (Figure 4) and mobile-first consumption (57.6% via iPhone or Android devices) emphasized the importance of cross-platform accessibility in science communication. Our podcast series demonstrated consistently high listener engagement, providing an open and accessible platform for scientific exchange across the global MR community. The high engagement among young listeners—particularly in countries like the US, UK, Germany, the Netherlands, and Japan—may reflect both a generational shift in media consumption for knowledge and skills improvement and the presence of well-established MR research centers that foster curiosity and ongoing learning. Moving forward, we envision enhancing our recording quality using professional audio equipment, exploring multilingual episode formats, and integrating podcast interviews with conference abstracts and featured presenters to further amplify scientific dialogs. Our podcast demonstrates the potential of audio-based communication to enrich education, inclusion, and collaboration in the field of MR.
Moss ZHAO (Stanford, USA), Daniel HOINKISS, Melanie BAUER, Hendrik MATTERN, Sanam ASSILI, Patricia CLEMENT, Joana PINTO
14:00 - 15:30
#45756 - PG370 Systematic Review of the role of Machine Learning in neuroimaging of pediatric brain tumors.
PG370 Systematic Review of the role of Machine Learning in neuroimaging of pediatric brain tumors.
Artificial Intelligence, which can be used to solve tasks. Recently, there has been an exponential growth in publications using ML in medicine, but the quality of the articles is heterogenous. Thus, several quality scoring tools have been created over the years to assess quality of these articles using ML. The METhodological RadiomICs Score (METRICS) is one of them and it is the latest quality scoring tool to have been made available. Aim of this research is to perform a systematic review of the studies using radiomics and ML in neuroimaging of PPBT and to evaluate their methodological quality with the METRICS score.
Four radiologists performed systematic research of the scientific literature on Pubmed, Scopus and Web of Science including articles up to October 2024. Methological quality was assessed with the METRICS score [1]. In total, 361 articles were found, but only 51 of them were eligible considering the inclusion criteria, which were : population aged less than 19 years of age, original research articles, and use of radiomics, ML and/or Artificial Intelligence in neuroimaging of PPBT (fig. 1). The biggest part of the included articles was multicentric (41%) and used manual segmentation (58.8%). The METRICS score was moderate in 47% of cases with the highest score being 77 and 29.4 the lowest (fig. 2). Our study has some limitations. Firstly, the studies included are retrospective. Secondly, MRI protocols are heterogeneous. Plus, some articles did not use the WHO 2016 classification to classify PPBT [2]. Finally, the METRICS score is relatedly new thus requires further validation. ML has shown multiple promising applications in neuroimaging of PBT [3]. Although the overall quality of the articles in this field is moderate, it needs to be implemented to use these tools in clinical practice.
Teresa PERILLO (Naples, Italy), Claudia GIORGIO, Mattia SICA, Andrea PONSIGLIONE, Arnaldo STANZIONE, Lorenzo UGGA, Gaetano UNGARO, Salvatore LAVALLE, Carmine FRASCA, Renato CUOCOLO
14:00 - 15:30
#45628 - PG371 Impact of Radiofrequency (RF) Coil Design on the Diagnostic Performance of Sodium (²³Na) MRI for Tissue Characterisation: Clinical Research Evidence.
PG371 Impact of Radiofrequency (RF) Coil Design on the Diagnostic Performance of Sodium (²³Na) MRI for Tissue Characterisation: Clinical Research Evidence.
Sodium (23Na) MRI at ultra-high field (UHF) (B0 ≥7 T) enables non-invasive mapping of tissue sodium concentration (TSC), providing insights into cellular viability and ion homeostasis, with elevated sodium levels commonly observed in malignant tumours compared to normal or benign tissues [1][2][3]. However, various technical challenges remain—including low signal-to-noise ratio (SNR), rapid biexponential T₂ decay, and strong partial volume effects (PVEs)—hinder its clinical translation [4][5]. A variety of RF coil designs are available, such as surface coils, birdcage volume coils, single/dual-tuned coils, and phased array receive coils, with each offering different trade-offs in SNR and spatial resolution. This systematic review evaluates how different RF coil designs affect the diagnostic performance of quantitative ²³Na MRI based on clinical evidence.
A systematic review following PRISMA 2020 guidelines was conducted (Figure 1) [6]. PubMed was searched (2004–2024) using a structured string combining medical subject headings (MeSH) terms and keywords related to 23Na MRI and pathology. Studies were included if they quantitatively assessed TSC in clinical populations, reported means and standard deviations, and involved at least five participants per group. Extracted data included organ system, field strength, RF coil type, and TSC measurements. TSC contrast reflects the absolute difference in TSC between malignant and beningtissues, while effect size (d) is the standardised magnitude of the difference between separation of of malignant and bening tissue using TSC. Bar charts visualised performance differences across coil designs, grouped into surface coils, birdcage coil, knee-coil, clamshell coil, and more with respect to effect size (d) (Figure 3). Fifteen studies were included, covering breast, kidney, prostate, and cardiac tissues, at various field strength (Figure 2). TSC contrasts ranged from 0.5 mM to 66.3 mM, with effect sizes ranged from 0.25 to 3.64. Clear separation (d > 0.8) was achieved in 9 studies, while others exhibited overlap between normal and pathological tissues (Table 1). For instance, Zaric et al. [7] demonstrated clear separation in breast cancer using a surface coil (d = 3.30), whereas Horvat-Menih et al. [8] and Barrett et al. [9] reported moderate separability in renal (d = 1.47) and prostate (d =1.09) imaging, respectively. Studies using multi-channel phased arrays for signal reception showed good performance with enhanced TSC contrast. Single-tuned sodium coils generally provided better performance than dual-tuned coils, while volume coils, such as birdcage, offered good B₁ homogeneity but lower sensitivity. RF coil design was a dominant factor influencing the diagnostic accuracy of ²³Na MRI. The use of multi-channel phased arrays and/or single-tuned coils enhanced SNR and this leads to increased spatial resolution and improved tissue characterisation. Volume coils and dual-tuned coils exhibited lower sensitivity, which reduced the effectiveness of TSC-based tissue differentiation. Additional factors affecting coil performance included PVEs, tissue heterogeneity, and sequence optimisation [10]. These factors could be improved the use of independent proton coils for tissue localisation and B0 shim corrections. For sodium imaging, the use of volume transmit coils, which are particularly effective in achieving B₁⁺ homogeneity in large anatomical regions such as the torso combined with phased array receive coils to enhance sensitivity, will be essential for realising the diagnostic potential of ²³Na MRI in oncology, nephrology, and cardiology. This review highlights that multi-channel phased surface arrays and the use single-tuned rather than double-tuned sodium coils significantly improve diagnostic performance in ²³Na MRI. Future RF coil developments could emphasis higher channel densities, flexible conformal surface arrays, advanced decoupling, and potential integration of volume Tx coil with phase Rx array in sodium combined with proton coil arrays, will further enhance clinical translation, supporting early disease detection and more personalised treatment strategies.
Un Hou CHAN (London, United Kingdom), Antoine NAEGEL, Sarah MCELROY, Vicky GOH, Özlem IPEK
14:00 - 15:30
#47313 - PG372 Quantitative MRI Techniques for Detecting Type 2 Diabetes-Related Brain Changes: A Systematic Review.
PG372 Quantitative MRI Techniques for Detecting Type 2 Diabetes-Related Brain Changes: A Systematic Review.
Type 2 Diabetes Mellitus (T2DM) is a prevalent and chronic metabolic disorder marked by high blood glucose level and insulin resistance. T2DM patients may experience cognitive impairments in areas such as executive function, memory, attention and visuospatial skills. Such cognitive changes are hypothesized to result from altered cerebral blood flow (CBF) and increased iron deposition in the brain.
As such, advanced quantitative MRI techniques have emerged as essential tools to investigate the underlying neural mechanisms and potential biomarkers of cerebral dysfunction. This review focuses on studies involving the evaluation of the sensitivity and specificity of selected quantitative MRI techniques namely Arterial Spin Labelling (ASL), Quantitative Susceptibility Mapping (QSM), and T1/T2 relaxometry in detecting diabetes-related cerebral alterations.
Using PubMed, a systematic search was carried out to identify human studies in the English language involving investigations of the impact of T2DM on the brain through advanced quantitative MRI techniques.
The search query used was: ("Diabetes Mellitus, Type 2"[MeSH] OR "Type 2 Diabetes Mellitus" OR "T2DM" OR "Type 2 Diabetes") AND ("Brain"[MeSH] OR "Cerebral" OR "Neuroimaging" OR "White Matter") AND ("Multiparametric Neuroimaging" OR "Quantitative MRI" OR "Quantitative Susceptibility Mapping" OR "QSM" OR "T1 mapping" OR "T2 mapping" OR "Arterial Spin Labelling" OR "ASL" OR "T1 relaxometry" OR "T2 relaxometry" OR "Relaxation time measurement")
The earliest published paper from the search was from 2017. As a result, no strict date range was applied to the search period. Relevant earlier works cited in the included papers were also considered.
The inclusion criteria required that studies: primarily investigate T2DM; include at least 10 participants per group; and utilize quantitative MRI methods. Reviews were excluded.
The search resulted in14 research articles, which were subsequently analyzed using the PICO framework. ASL-based perfusion imaging was the most frequently used MRI technique. However, studies assessing whole-brain CBF using solely ASL produced inconsistent results across different studies. To address this issue, ASL was often combined with fMRI, leading to more consistent insights particularly when calculating metrics such as the regional homogeneity (ReHo) to CBF ratio. Findings from these studies indicated region specific hypoperfusion in T2DM patients, notably in areas associated with memory and executive function, such as the hippocampus, precuneus, and posterior cingulate cortex.
QSM was used in three studies to quantify brain iron deposition. Increased susceptibility values were observed in regions like the putamen, substantia nigra and Parahippocampal gyrus in T2DM patients. This was significantly visible on those with mild cognitive impairment.
One multimodal study combined structural MRI, diffusion tensor imaging (DTI), ASL, resting-state fMRI, FDG-PET, and retinal OCT to assess the effect of T2DM on brain structure and function. The study found reduced attentional performance, lower nucleus acumens volume, and decreased cerebral glucose metabolism in individuals with T2DM, despite no significant differences in cerebral perfusion or white matter microstructure.
Notably, no studies were found utilizing T1 and T2 relaxometry. While CBF results derived from ASL alone were inconsistent, several studies emphasized the importance of multimodal MRI approaches in retrieving early signs of neural compromise in T2DM. Neurovascular Coupling (NVC) measures, derived from combining ASL with fMRI, showed stronger associations with cognitive performance and disease duration, offering a more detailed picture of how T2DM alters brain function. Notably, the integration of ASL with fMRI introduces non-quantitative measures, such as ReHo; nevertheless, this combination remains valuable for uncovering patterns of disrupted NVC. Studies also highlighted that voxel-based and region-specific analyses provided greater sensitivity than global measurements, uncovering localized abnormalities that whole-brain approaches might miss.
QSM studies introduced iron deposition as an important mechanism of cognitive dysfunction, aligning with known pathways of oxidative stress and inflammation in T2DM. The relationship between elevated iron levels and impaired cognition, particularly executive dysfunction and memory loss, supports the use of QSM in monitoring disease progression and therapeutic response. Quantitative MRI techniques especially ASL, fMRI, and QSM demonstrate significant promise in detecting cerebral alterations in T2DM patients. Multimodal imaging approaches and advanced analytical techniques such as voxel-wise and region-specific assessments enhance diagnostic sensitivity, potentially allowing earlier detection. Altered perfusion, iron deposition, and cognitive decline highlight neuroimaging biomarkers’ role in T2DM related brain dysfunction.
Nolan VELLA (Malta, Malta), Claude Julien BAJADA, Matthew GRECH SOLLARS, Carmel J CARUANA
14:00 - 15:30
#47796 - PG373 Infection Prevention and Control in Magnetic Resonance Imaging: A Scoping Review of Current Evidence and Practice.
PG373 Infection Prevention and Control in Magnetic Resonance Imaging: A Scoping Review of Current Evidence and Practice.
Magnetic Resonance Imaging (MRI) is a critical diagnostic modality that presents unique challenges for infection prevention and control (IPC) due to its strong magnetic field, non-ferromagnetic equipment requirements, and access-controlled environment [1]. A recent scoping review of IPC in medical imaging departments indicated that IPC education would benefit from a systems approach and further research [2].. The necessity of IPC within healthcare settings is universally acknowledged as critical to providing safe and high-quality care [3]. This scoping review aims to map the existing literature and identify the gaps in research concerning IPC practices within MRI. By examining the range and nature of studies, including guidelines, empirical research, and expert opinions, this review will provide a comprehensive overview of the current knowledge on the specialised IPC measures required in MRI suites. It will highlight the challenges and limitations of current practices around the world, outline the specific needs and considerations for effective IPC in MRI settings, and suggest areas for future research and policy development.
A scoping review methodology was employed in accordance with the Joanna Briggs Institute framework [4]. A comprehensive literature search was carried out utilising several electronic databases, including Medline, Scopus, CINAHL, Web of Science, and Google Scholar, to pinpoint relevant peer-reviewed articles. The search concentrated on topics pertaining to IPC within MRI settings. Searches were restricted to articles published in English from 2000 to 2024, with a focus on healthcare environments that utilise MRI and associated clinical workflows. Covidence was employed for data management and screening [5], while the SPIE [4] framework facilitated structured evidence synthesis. The PRISMA checklist was utilised to ensure transparent and efficient reporting [6]. Ultimately, a total of 1551 articles were identified; of these, 49 progressed to full-text review, and through further screening, 27 studies were included in the review. The review encompassed 27 studies, predominantly concentrating on guidelines and recommendations for infection prevention and control (IPC) in medical imaging departments. A notable surge in published articles was observed in 2020. Of the 27 articles evaluated, 17 pertained to COVID-19 IPC, with only 4% specifically addressing MRI, primarily focusing on imaging-related services during the COVID-19 pandemic. Additionally, six articles were associated with investigative studies; one emphasised that patient contact items ranked among the most contaminated surfaces within MRI suites [7]. The four narrative reviews examined either hospitals or imaging departments that had established IPC guidelines, yet none specifically addressed IPC in the context of a magnetic field [2,8,9,10]. This review highlights the lack of MRI-specific studies in relation to IPC. Many studies mentioned MRI as part of the medical imaging department but there was a lack of specific interventions and guidelines dedicated to the MRI environment. The intricacies of MRI environments and the vulnerability of staff and patients to infections necessitate specialised IPC skills and practices tailored for radiographers and other personnel operating within these settings. Despite the critical role of IPC in healthcare, there is a noticeable deficit in evidence-based guidelines specifically addressing the needs and challenges faced in MRI suites.
Frances GRAY (Sydney, Australia), Yobelli JIMENEZ, Dania ABU AWAAD, Sarah LEWIS, Peter KENCH
14:00 - 15:30
#46104 - PG374 Resting State fMRI: Implementation Of A Quality Control Pipeline In The WELDFUMES Study.
PG374 Resting State fMRI: Implementation Of A Quality Control Pipeline In The WELDFUMES Study.
Functional MRI (fMRI) relies on blood oxygenation level-dependent (BOLD) signals, where neural activity–driven changes in oxygen consumption alter MR signal intensity. However, BOLD data are susceptible to neural and non-neural noise sources, head motion, and limitations of echo planar imaging [1]. Quality control (QC) is therefore needed to ensure data integrity and MR acquisition parameter consistency, particularly in multisite studies. This study developed a QC workflow combining qualitative and quantitative methods to assess resting-state (rs) fMRI data from a cohort of Swedish welders exposed to varying levels of manganese-containing welding fumes [2]. The aim was to use quantitative metrics to detect extreme outliers and to improve the overall data quality by minimizing exclusions.
Anatomical and rs-fMRI data were from the WELDFUMES study [2], involving 51 healthy male welders (ages 21–63) scanned at two sites using three 3T scanners: GE MR750w, GE Signa Premier, and Siemens Prisma. Imaging included T1-weighted fast gradient inversion recovery [2] and an EPI sequence (TR/TE: 1.5–2.5 s/30 ms; temporal resolution: 315–236 ms; voxel dimensions: 64×64 or 128×128) using 24", 48" and 64" channel head coils.
Preprocessing of rs-fMRI and anatomical data, including realignment, unwarping [3, 4], and slice timing correction [5, 6], was performed using the CONN toolbox (RRID: SCR_009550, v22.v2407.a) [7,8] and SPM12 (RRID: SCR_007037, v12.7771) [9]. Volumes with frame-wise displacement >0.5 mm or BOLD signal >3 SD were classified as outliers [10, 11]. The outlier-excluded mean BOLD and anatomical data were normalized, co-registered into the standard MNI space, segmented into GM, WM, and CSF, smoothed (8 mm FWHM Gaussian kernel), and resampled (2 mm isotropic) [12, 13]. The default denoising pipeline in CONN [15] regressed out confounds (WM/CSF, motion parameters, and outlier scans), followed by high-bandpass filtering of BOLD signals above 0.01 Hz.
Visual and automated QC was applied to the rs-fMRI and anatomical datasets: raw data QC assessed subject demographics, MR acquisition parameter consistency, and image quality; preprocessed data QC determined accuracy in segmentation, normalization, co-registration, and potential artifact; and denoised data QC evaluated the impact of residual noise on the distribution of functional connectivity. Sample-specific threshold values of Q3 + 3IQR or Q1 – 3IQR were used to identify extreme outliers [1]. One subject was excluded due to missing fMRI data. The fMRI sequence protocols differed in temporal resolution (TR = 1.5 or 2.5 s; 236 or 315 measurements) and spatial parameters (voxel sizes 3.4 x 3.4 x 3.7 mm³ or 3.8 × 3.8 × 3.6 mm³). Differences in RF sensitivity maps of the head coils (24", 48", and 64") may have contributed to signal variability in the BOLD data from the 48" and 64" coils, as shown in Fig. 1B. The effect of incorrect orientation on preprocessing is illustrated in Fig. 2.
The results of the automated quantitative QC are shown in Fig. 3, where the metric Norm-Struct confirmed Subject 3's anatomical images as an extreme outlier due to incorrect slice orientation. In contrast, no extreme outliers were detected in the normalized (Norm-Func) and denoised (DOF) fMRI data; however, Subject 19 had 76 outlier scans, and a high MeanMotion was found in Subjects 19 (mean = 0.40) and 41 (0.41). Fig. 4 shows the distribution of functional connectivity (FC) and QC-FC correlations before (top) and after (bottom) denoising. FC was markedly biased and highly variable (mean correlation r = 0.23, SD = 0.305) but shifted closer to zero with reduced variability (r = 0.02, SD = 0.15) after denoising. The QC-FC association (r) had a mean = 0.03 (77% matches), SD = 0.13, which reduced to mean = 0.00 (88 % matches), SD = 0.16 after denoising. In this study, we implemented an automated QC pipeline suitable for multisite MR data. Combining qualitative and quantitative methods, we assessed rs-fMRI data quality from the WELDFUMES project. Metrics such as Norm-Struct, Norm-Func, and visual inspection identified artifacts, acquisition inconsistencies, and evaluated key preprocessing steps including normalization, segmentation, and co-registration to MNI templates.
Given the impact of motion and subject-specific factors on the BOLD signal, quantitative metrics such as MeanMotion, PVS, DOF, and FC distributions were used to improve the reliability of the fMRI data. QC-FC was then used to measure the association between functional connectivity and residual noise. Extreme outliers were defined using sample-specific thresholds, calculated as Q3 + 3 × IQR or Q1 – 3 × IQR [1]. A few subjects were in the extreme outlier zones (Norm-Struct, MeanMotion, and PVS, Fig. 3); however, there were no additional exclusions after denoising (DOF, Fig. 3). In summary, the implemented QC workflow was highly effective at evaluating rs-fMRI data quality, and this approach could be adapted for task-based fMRI.
Mary ADJEIWAAH (Linköping, Sweden), Per THUNBERG, Gunilla WASTENSSON, Göran LIDEN, Bernt BERGSTRÖM, Louise FORNANDER, Peter LUNDBERG
14:00 - 15:30
#47924 - PG375 Dynamic Brightness and Contrast Adjustment for Enhanced Visualization of Medical Images in PowerPoint Presentations.
PG375 Dynamic Brightness and Contrast Adjustment for Enhanced Visualization of Medical Images in PowerPoint Presentations.
In medical imaging, accurate and clear visualization of diagnostic images is crucial for clinical decision-making, communication, and education. Magnetic Resonance Imaging (MRI) and similar modalities often rely on high-resolution images that may lose diagnostic value when displayed under poor lighting or with fixed contrast settings during presentations. This project addresses the limitations of static image enhancement by developing a real-time, user-controlled brightness and contrast adjustment tool integrated directly into Microsoft PowerPoint.
The developed tool utilizes Visual Basic for Applications (VBA) scripting within Microsoft PowerPoint, combined with Windows API functions,
to allow users to adjust images of the current slide regarding brightness and contrast dynamically based on mouse movements and keyboard
commands.
The process begins when the user presses the "M" key, which triggers the monitoring. The program initializes the mouse position (startX and startY)
and sets the variables isTracking = True and = False to start tracking mouse movements. The main loop then calculates the horizontal (deltaX) and
vertical (deltaY) mouse movement to adjust brightness and contrast accordingly. As long as isPaused remains False, the adjustments continue. The
brightness and contrast values are constrained within a range of 0 to 1. The program iterates through all shapes on the slide, checking if they are
images (msoPicture). If so, the brightness and contrast are adjusted. Additionally, several key presses are monitored: "S" pauses the adjustments
(isPaused = True), "R" resumes them (isPaused = False), and "X" resets all settings to their original values. Finally, the DoEvents function ensures the
program responds to system events without freezing. The Program flow chart is shown in figure 2. The tool enabled smooth, real-time image enhancement during PowerPoint presentations. It performed consistently across different medical image formats, including MRI, CT, and X-ray scans. Users were able to intuitively modify image properties mid-presentation, with immediate visual feedback. Enhanced visibility of fine details—such as tissue structure, lesions, or vascular patterns—was achieved, especially under suboptimal lighting. Presenters could adapt visual settings based on the audience and context, improving the clarity and effectiveness of medical communication. The implementation proved to be robust and user-friendly. It required no prior image editing or additional software, making it practical for clinical, academic, and conference settings. The system allowed for dynamic, responsive control over image display without interrupting presentation flow. Feedback from test users highlighted ease of use, flexibility, and increased confidence in image-based explanations. The tool’s performance remained stable regardless of image resolution or presentation environment. This project successfully introduced an intuitive, real-time brightness and contrast adjustment tool for PowerPoint-based medical image presentations. By leveraging native VBA capabilities and API functions, the solution improves diagnostic clarity without the need for external software. Future improvements could include window level/width adjustments for radiology applications and better support for high-resolution or compressed image formats. With continued refinement, the tool could become a standard feature for medical educators and clinicians aiming to enhance visual impact in digital presentations.
Amira ALOUANE, Helena NAWRATH, Amira ALOUANE (Hagen, Germany)
14:00 - 15:30
#45959 - PG376 Spoiled, Spoiled or Spoiled?
PG376 Spoiled, Spoiled or Spoiled?
Throughout MR literature, often an ambiguity arises with the term “spoiled” or “spoiling”. In the generic field of gradient-echo sequences (mostly abbreviated as “GRE” or “FFE”)[1,2], “spoiling” usually refers to measures aimed to reduce unwanted transverse magnetization. However, the sequence can be “spoiled” (or sometimes “crushed”) according to – at least – three different variants:
- RF-spoiled, which involves phase-modulating the excitation pulses[3] by n^2 ϕ_0, where n is the pulse number and ϕ_0 the golden angle or some other optimized angle, which may be 117 or 150 degrees.
- Incoherent-gradient-spoiled, where a net gradient area of varying size is applied between successive RF pulses. Although the use of this type of spoiling is somewhat archaic, it cannot be ruled out that “spoiling” refers to this spoiling variant.
Coherent gradient echo applies a constant nonzero gradient area between any pair of successive RF pulses. According to some definitions[1,3], this variant is not to be called “spoiled” at all, since it does preserve transverse coherence, as do balanced sequences. Nevertheless, many papers do call this choice “spoiled” (see an incomplete but representative literature review in the supplementary material).
Many acronyms for spoiled sequences are available, but these can be ambiguous on their own, so many authors just publish their sequence as “spoiled”. It is the purpose of this abstract to quantify the occurrence of adequate specification of this term.
A set of publications were selected to examine the specification of the word “spoiling” (or “spoiled”). The was selection that was primarily drawn from references used in my PhD thesis and underlying publications[4–6]. A further selection criterion was the presence of the string “spoil” (as in spoiling, spoiled, spoiler) in the context of gradient-echo sequences. Hits in references were not considered. This delivered over 80 hits. By a random selection, the set was further limited to 50 hits.
In these publications, the string “spoil” was examined on whether
• It clearly specified the type of spoiling (e.g. by mentioning “RF-“, “Incoherent gradient-“ or “Coherent gradient-“ in the immediate vicinity of the word “spoiling”) or the immediate context specifies the type of spoiling.
• It mentions “gradient spoiling”, without clarifying whether it is coherent or incoherent.
• It is specified as “SPGR”, which may be RF-spoiling or incoherent gradient-spoiling.
• Or it just mentions “spoiling”, leaving the reader to search through the context to guess which type of spoiling is used.
The “immediate context” was defined as within one page of the first occurrence of the word “spoiling” (or “spoiled”). Details of the list of publications can be seen in supplementary material (https://doi.org/10.5281/zenodo.15201724). The results are visualized in Figure 1. Although the MRI-world is not really scarce on acronyms, there is no straightforward and unambiguous acronym for the coherent gradient-echo case. “FFE” (the Philips-entry in the MR-TIP[2] table) is clearly ambiguous, since “FFE” is also used as an umbrella-term for all types of gradient-echo sequences. Also “Spoiled FFE” is ambiguous, since it also can refer to RF-spoiled gradient-echo. “FISP” might be an unambiguous acronym, but this term is somewhat troubled by the presence of the technology called “trueFISP” (balanced, i.e. zero net gradient area between successive pulses). Is “FISP” then an umbrella term encompassing “trueFISP” as well as something like “untrue FISP”, or does “FISP” just refer to the latter? Is “FISP” referring exclusively to the “untrue” version of itself?
“SSFP” (Steady-State Free Precession) might be another candidate for an unambiguous term for a sequence using coherent gradient spoiling. But it isn’t: although the one-time Shimadzu acronym referred indeed to coherent gradient spoiling, the MR-TIP table puts the GE-term “SSFP” in the same row as the Siemens-term “PSIF” (a Siemens-term referring specifically to the echo before the RF-pulse in a coherent gradient spoiling sequence); then it adds to the confusion by stating “A new family of steady state free precession sequences use a balanced gradient (…)” – which seems to make “SSFP” also an umbrella term.
Such ambiguities might cause researchers to avoid acronyms altogether and specify their sequence as “spoiled”. Yet, as shown, 38% of publications insufficiently specify the type of spoiling. When discussing gradient-echo MR sequences, the word “spoiled” (or “spoiling”) should not be used in isolation; these terms should be padded with either “RF”, or with “incoherent gradient-“, or with “coherent”. The latter (i.e. coherent spoiling, or coherent gradient-spoiling) should be read as “Gradient-echo sequence with a constant unbalanced gradient moment in each TR”. Of course, it is equally valid to not call this “spoiling” at all[1,3].
Miha FUDERER (Utrecht, The Netherlands)
14:00 - 15:30
#47808 - PG377 How to set acquisition parameters for accurate quantification of magnetic susceptibility? Application to Parkinson's disease.
PG377 How to set acquisition parameters for accurate quantification of magnetic susceptibility? Application to Parkinson's disease.
One of the hypotheses explaining the origin of Parkinson's disease (PD) is the abnormal deposition of iron in specific deep gray nuclei (DGN) [1]. In vivo quantification of iron from magnetic susceptibility (χ) maps obtained by quantitative susceptibility mapping (QSM) could provide an early neuromarker of PD.
In this clinical context, optimization of acquisition parameters (number of echoes, spatial resolution, bandwidth) is important for robust χ quantification with reduced acquisition time [2].
Phantom: Tubes containing solutions of Dotarem® (Gadoterate Meglumine; 0.5 mmoL.ml-1; Guerbet; France) (Fig. 1) at various concentrations of known χ [3].
Healthy volunteers: Thirty-two healthy volunteers (HV) with no history of neurological disorder were recruited from 2021 to 2024 (Clinical trial NCT05107232; Univ. hospital of Rennes) and divided into two age groups (group 1: n=16; sex ratio=1; age=22±1 years and group 2: n=16; sex ratio=1; age=49±2 years).
MRI data acquisition: QSM data were acquired at 3 T (Magnetom Prisma VE11C; Siemens Healthineers; Erlangen; Germany) using a 64-channels head coil. For phantom and HV, a 3D monopolar susceptibility weighted imaging (SWI) multi-gradient-recalled-echo (MGRE) sequence was used. Several parameters were evaluated such as the number of echoes, TR, TEmin/ΔTE, spatial resolution, and bandwidth. All acquisition parameters are resumed in Table 1. Acquisitions on phantom were repeated five times, during different MRI sessions. For HV, the protocol included 3D anatomical T1 and T2 weighted anatomical images (T1w and T2w) with a spatial resolution of (1 mm)3 to segment the DGN of interest.
Image processing: Reconstructions of χ maps were computed on MATLAB (v2017Rb) using Sepia (v1.1.1). For phantom, the FSL brain extraction tool was manually settled to adapt it to the geometry of the tubes and three processing pipelines were evaluated [4]. The ImageJ (v1.53g; National Institute of Health) software was used to quantify the mean χ values and standard deviation (SD) of each manually traced ROI on χ maps. For HV, phase unwrapping, background field removal and field to susceptibility inversion were processed using an in-house developed processing pipeline based on ROMEO, PDF and MEDI algorithms [4] to calculate χ maps (Fig. 1, Fig. 2-A). In addition, six regions per hemisphere were segmented from the T1w image using the recon-all process of FreeSurfer [5] (v7.2.0) (caudate nucleus (CN), putamen (Put), globulus pallidus (GP)), and from the T2w image using pBrain [6] (substantia nigra (SN), red nucleus (RN) and subthalamic nucleus (STN)). The mean χ values and SD of each DGN were quantified using 3D Slicer (v5.6.2).
Statistical analysis: For phantom, a non-parametric statistical test was performed to compare χ quantification between the acquisition parameters. For HV, a parametric statistical test was performed to compare χ values quantified in the DGNs between the two groups. On phantom, increasing the spatial resolution involved an underestimation of the χ, due to the loss of signal-to-noise ratio, while reducing the number of echoes enabled reliable quantifications until five echoes. These observations led us to concentrate our in vivo acquisitions on the MGRE sequences up until five echoes. For HV, reducing the number of echoes from eight to five gave a quantification consistent with the literature [7] (Fig. 2-B), and no statistically significant difference was observed in function of the number of echoes used except in the group 2 for CN (8 vs 6 echoes and 8 vs 5 echoes: p<0.05). The aging effect was observed for the protocol using height, six and five echoes and was statistically significant for five DGN out of six (CN, Putamen, GP, RN: p<0.01; STN: p<0.05) demonstrating the robustness of a five-echo MGRE sequence in vivo. In vitro, reducing the number of echoes from eight to five not impaired χ quantification. However, we demonstrated in vitro that a SWI MGRE sequence with only four echoes did not produce accurate and reproducible χ quantification within the ranges targeted in the DGN. In vivo, changing the spatial resolution from (1.5 mm)³ to (1 mm)³ not improved χ quantification. However, reducing the number of echoes to five allowed us to obtain a robust χ quantification consistent with the literature [7], while still enabling the detection of aging effect. These optimizations allowed us to reduce the acquisition time from 6mins31s to 3mins48s. The sequence is ready for inclusion in a multiparametric clinical protocol, with a spatial resolution of (1.5 mm)³. These results make it possible to apply the MGRE with five echoes acquisition protocol on PD patients for in vivo quantification of χ in DGN, to explore the potential of this new neuromarker for monitoring PD progression and identifying various profiles of patients.
Aurélien HERVOUIN (Rennes), Pierre LEMOIS, Guy PECHEUL, Johanne BEZY-WENDLING, Fanny NOURY
14:00 - 15:30
#47922 - PG378 4D MR spirometry across magnetic field strengths: a multicentric travelling healthy volunteer study at 0.55, 1.5 and 3T.
PG378 4D MR spirometry across magnetic field strengths: a multicentric travelling healthy volunteer study at 0.55, 1.5 and 3T.
Lung MRI has long been constrained by intrinsically low proton density, short T2* relaxation times—limitation that is enhanced at high magnetic field strengths making it challenging [1]. Recent advances in ultra-short echo time (UTE) sequences and self-navigated respiratory gating now enable time-resolved, 4D pulmonary (3D + time) MRI in free-breathing conditions [2,3]. In this context, low field MRI systems (<0.55T) offer a trade-off between SNR and T2* relaxation [4]. Also, lower requirements for field homogeneities allow wider bores enhancing accessibility for claustrophobic or dyspneic patients.
This study investigates the impact of magnetic field strength on image quality and dynamic pulmonary function assessment, using the exact same MR sequence. We hypothesize that the trade-offs inherent to field strength—particularly regarding signal-to-noise ratio and relaxation properties—can be quantified and exploited to optimize MRI-based lung imaging, with the potential of low-field systems for reproducible, radiation-free, region-specific imaging in free-breathing conditions.
Lung MRI was performed at 0.55T, 1.5T, and 3T (Free.Max, Sola, Skyra; Siemens-Healthineers) using a custom dynamic center-out radial UTE spoiled GRE sequence [5] with AZTEK trajectory sorting [3] and self-navigated respiratory gating. To ensure intra-subject comparability, the same healthy volunteer was scanned across three different clinical sites.
The UTE sequence consists of a center-out radial acquisition lasting 11min with: TR/TE/flip angle=3ms/0.03ms/4°, FOV 32x32x30cm3, voxel size (2mm)3, matrix size [160,160,150]. The maximum gradient amplitude and slew rate were constrained by the lower hardware performance of the 0.55T system (15mT/m and 40T/m/s). Retrospective respiratory gating and image reconstruction were computed for 32 respiratory gates [6].
To assess the impact of magnetic field strength on image quality, respiratory-resolved images were evaluated based on the following criteria: the visibility and spatial extent of the pulmonary vascular tree and the contrast-to-noise ratio (CNR). CNR is computed as the ratio between lung-liver signal and the noise.
To enable future inter-subject comparison, accurate lung segmentation is needed. Segmentation quality relies on sufficient CNR and clear delineation between lung parenchyma and adjacent muscular structures. Special attention was given to the diaphragm, whose substantial respiratory motion makes it especially challenging to segment consistently. The 4D MRI acquisition also provided full diaphragmatic coverage across the respiratory cycle, allowing dynamic segmentation required for registration and subsequent comparative analysis. The visualization of the pulmonary vascular tree was assessed using MIP over 2 cm sagittal plane (Fig. 1). At both 0.55T and 1.5T, vessels were clearly visible up to the second generation, with well-defined contours and consistent parenchymal contrast. In comparison, at 3T, vascular structures appeared with lower contrast, and vessel contours were less defined.
Qualitative assessment of the lung–diaphragm interface (Fig. 2) revealed notable differences in anatomical sharpness across field strengths. The signal profiles were sharply delineated at 1.5T, moderately at 0.55T and blurred at 3T.
Quantitative analysis of CNR further supported these findings with the highest CNR observed at 1.5T CNR₁.₅=34, followed by 0.55T, CNR₀.₅₅=20, and lowest at 3T, CNR₃=4.9.
To evaluate dynamic image consistency, respiratory-resolved images were analyzed across multiple gates (Fig. 3). The spatial distribution of normalized signal intensity remained stable across the respiratory cycle for all systems. However, minor motion blur was observed in gates corresponding to phases of high airflow particularly at 3T. Our findings demonstrate that magnetic field strength has a direct impact on 4D lung MRI quality. While 1.5T provided the best compromise between anatomical sharpness and signal stability, 0.55T preserved essential structural details despite a lower CNR. The poor results at 3T are non-satisfactory and could be due to technical limitations. This can lead to a less effective retrospective gating resulting in lower SNR and under sampling artefacts after reconstruction [7]. Improvements are underway to reduce the noise of the 0.55T and refine the reconstruction of the 3T system. In a travelling volunteer at 3T, 1.5T and 0.55T across three sites, 4D lung MRI was successfully reconstructed over 32 respiratory phases. Respiratory gating and anatomical delineation of the vascular tree and diaphragm were optimal at 1.5T and very encouraging at 0.55T. These findings support the potential of low and intermediate fields with ongoing developments aimed at optimizing low-field acquisition and reconstruction.
Timothee CAUSSIN (Courbevoie), Alexiane PASQUIER, Anna REITMANN, Samia BOUSSOUAR, Aurelien MASSIRE, Naila BOUDIAF, Rose Marie DUBUISSON, Xavier MAITRE, Angeline NEMETH, Marie POIRIER QUINOT
14:00 - 15:30
#47944 - PG379 Evaluating methods for scanner harmonisation in T1w MRI (Cam-CAN Study).
PG379 Evaluating methods for scanner harmonisation in T1w MRI (Cam-CAN Study).
MRI data often lacks robustness due to variability introduced by site, scanner type, and hardware/software changes over time [1,2,3]. This variability can impact downstream analyses of brain structure, including measures of healthy tissue or lesion volumes [4,5]. The Cam-CAN dataset is a longitudinal, multi-modal, single-site study collected over 12 years, during which two major hardware changes occurred: a gradient coil replacement and a scanner upgrade (PRISMA vs. TRIO) [6]. We evaluated the robustness of Cam-CAN T1w structural data against these changes and whether pre- and post-processing techniques reduced effect sizes.
Two different datasets were used to evaluate hardware changes and FreeSurfer was used for cortical parcellation of 68 ROIs, with visual quality assessment and outlier detection. Inter- and intra- canner differences were assessed in a Traveling-Heads (n=20) repeated measures dataset of 5 patients each scanned twice on both TRIO and PRISMA within two weeks. Skull stripped images were pre-processed using three time-based (Z-Score, White-Stripe, Kernel Density Estimation, Fuzzy Clustering) and 2 sample-based (Nyul histogram matching and Least-Square Means) SI normalisation techniques. Intensity comparisons were conducted using normalized overlap (intersection), Kullback-Leibler (KL) divergence, and the standard deviation of subject-level normalized mean intensity (SD NMIs). Effects on ROI thickness measures were compared using Coefficient of Variance (CoV), ANOVA, and paired t-tests (with Bonferroni correction). The gradient-coil change was assessed on 89 matched subject pairs (matched on sex, handedness, and age) and assessed the efficacy of NeuroCombat harmonisation in mitigating these coil-related biases. We first quantified coil-related effects in the raw data using paired t-tests and Cohen’s d (with FDR correction) across ROIs, both unadjusted and age-adjusted via linear mixed-effects models (LMMs). NeuroCombat-harmonised outputs were then subjected to the same analyses. We compared harmonised vs. original values using (1) scatterplots with Pearson r and RMSE, (2) Bland–Altman plots (bias ±1.96 SD). Scanner (TRIO vs PRISMA):
Across all inter-scanner comparisons, the observed overlap values ranged from 0.84 to 0.98 and maximum KL divergence observed was 0.06 (approximately a percent similarity of 94%) which indicated high overlap between distributions. No inter-scanner ROI differences were detected (p<0.05). We examined intensity differences across scanners (PRISMA vs. TRIO) by calculating the standard deviation of subject-level normalized mean intensity (SD NMIs) across all four scanning sessions. Significant scanner-related differences (p<0.05) were observed in repeated measures ANOVA and paired t-test comparisons in up to 27% of ROIs depending on the normalisation technique used. Non-normalised images demonstrated scanner related differences in 9.7% of ROIs. Nyul and LSQ pre-processed images affected 5.5% of ROIs, while other pre-processing techniques demonstrated an increased scanner variability ranging from 13% to 16%.
Gradient-coil change:
Coil replacement induced moderate biases (Cohen’s d range ≈ –0.30 to +0.28; 12/82 ROIs significant at FDR < 0.05), persisting after age adjustment (β-coil range ≈ –0.27 to +0.25; 9 ROIs significant). NeuroCombat compressed effect‐size distributions to near zero (d range ≈ –0.02 to +0.02; no ROI significant post–FDR), whether unadjusted or age‐adjusted. There was a strong preservation of inter-subject variance (median Pearson r > 0.97; RMSE < 2% of mean volume across sampled ROIs), minimal systematic offsets on Bland–Altman (mean bias < 0.5% of mean ROI value; limits of agreement ±3%). Scanner change introduce measurable shifts in raw intensities but had limited effects after FreeSurfer processing. The effects of pre-processing methods on ROI thickness measures were mixed, some introduced variability, while sample-based normalization techniques modestly improved scanner consistency. Although scanner-related differences were still detected in some analyses, their overall impact was limited. Gradient-coil–driven differences are not significant in FreeSurfer cortical thickness measures but NeuroCombat demonstrated a reduction in bias and compressed effect-size. These findings support its use in harmonising structural MRI data across scanner hardware changes in longitudinal and multi-site studies. Limitations of our study included a small traveling-heads sample. Given, the small size, it would be useful to further explore of sample-based SI normalisation techniques to reduce scanner variability in FreeSurfer derived cortical thickness measures. Future work could test multi-site generalizability of these techniques in combination with ComBat. Overall, our findings support the robustness of the Cam-CAN dataset across major hardware changes but suggest that intensity pre-processing could further enhance inter-scanner consistency in structural MRI analyses.
Tanvi RAO (Cambridge, United Kingdom)
14:00 - 15:30
#47787 - PG380 Assessing the role of ASL for accurate diagnosis of neurodegenerative diseases: a study in a memory clinic.
PG380 Assessing the role of ASL for accurate diagnosis of neurodegenerative diseases: a study in a memory clinic.
Dementia encompasses a group of clinical syndromes characterized by progressive decline in cognitive functions, most often caused by neurodegenerative diseases such as Alzheimer’s disease (AD), frontotemporal dementia (FTD), or Lewy body dementia (LBD). [1] Differentiating between these etiologies, particularly in the prodromal or early symptomatic stages, remains a clinical challenge. Neuroimaging has become essential for differential diagnosis, using structural MRI or [18F]-FDG PET. However, FDG PET involves exposure to radiation and a high cost together with limited availability. Arterial spin labeling perfusion MRI (ASL-MRI) appears to be a promising biomarker for the assessment of neurodegenerative diseases, providing a quantitative, non-invasive measure of cerebral perfusion. [2] However, its clinical adoption is hindered by lower signal-to-noise ratio and sensitivity to artefacts, and need to be more explored before clinical use. This study aims to compare the diagnostic performance of multiple neuroimaging modalities—structural MRI, FDG-PET, and ASL-MRI—in differentiating major neurodegenerative dementia syndromes in a clinical setting.
The study population included 78 patients with typical AD, 11 with behavioral-variant FTD (bvFTD), 8 with semantic-variant FTD (svFTD), 13 with LBD, and 15 healthy controls (HC). Imaging data—including 3D T1-weighted MRI, ASL-MRI, and FDG-PET—were acquired using a single GE Healthcare Signa 3T PET/MR hybrid scanner. Two ASL sequences were employed in the study: a single-delay 3D pCASL sequence and a multi-delay sequence incorporating seven distinct post-labeling delays.
Image preprocessing was conducted using SPM12 to obtain grey matter modulated probabilistic maps, FDG-PET scans and CBF maps in MNI space. Regions of interest (ROIs) were defined using 94 cortical regions from the AAL3 atlas [3], from which regional volumes, mean glucose metabolism, and mean cerebral blood flow were extracted. ASL sequence harmonization was performed at regional levels using the NeuroCombat algorithm. [4] Voxelwise group comparisons between patient subgroups and HC were conducted for each imaging modality using SPM12. Direct voxelwise comparisons between modalities were then carried out using W-score maps. [5] Covariates for all analyses included age, sex, total intracranial volume (for T1-MRI analyses) and sequence type (for ASL). Sparse Partial Least Squares Discriminant Analysis (sPLS-DA) was performed using the mixOmics R package [6] for three models: using regional volumes from T1-MRI alone (T1 model), combined with FDG-PET measures (T1+FDG-PET model), or with ASL measures (T1+ASL model) to classify AD, FTD (bvFTD and svFTD) and HC. Performance of the three models was evaluated using leave-one-out cross-validation and area under the curve (AUC) metrics to assess group discrimination. In AD, atrophy was pronounced in the medial temporal regions, especially the hippocampus (pFWE<0.05), while FDG-PET showed bilateral hypometabolism in the parietal regions, posterior cingulate, precuneus, medial temporal areas, and in the dorso-prefrontal cortex. ASL revealed perfusion deficits in the parietal and posterior cingulate regions but with much less intensity and extent. In bvFTD, MRI and FDG-PET demonstrated predominant frontal lobe abnormalities (puncor<0.001), which were not consistently captured by ASL. In the case of svFTD, all identified modalities revealed left anterior temporal abnormalities, with ASL changes exhibiting less marked severity (puncor<0.001). In LBD, no significant atrophy was observed (puncor<0.001), but FDG-PET revealed hypometabolism in the occipital, parietal, and temporal lobes; ASL showed limited left parietal perfusion reduction. Intermodality discrepancies were observed across syndromes. In AD, greater limbic atrophy contrasted with broader parieto-temporal hypometabolism and hypoperfusion. In bvFTD and svFTD, structural and metabolic abnormalities exceeded perfusion deficits. In LBD, hypometabolism was more widespread compared to other modalities. The ability to discriminate between groups was enhanced by the addition of either FDG-PET or ASL to T1-weighted imaging. However, the discriminative performance of the T1+FDG-PET model was comparable to that of the T1+ASL model. Despite less extensive abnormalities than FDG-PET, ASL showed similar spatial patterns and comparable classification performance, supporting its clinical relevance. [7-8] Limitations include diagnostic group imbalance, with AD predominance reflecting typical prevalence; ongoing recruitment aims to address this. Two ASL sequences were used, with statistical adjustments for quantification differences. Combined with structural MRI, ASL offers diagnostic accuracy comparable to FDG-PET in differentiating neurodegenerative dementias, supporting its potential as a non-invasive alternative. Further recruitment will clarify its clinical value across syndromes.
Mathilde NGUYEN (Paris), Nicolas VILLAIN, Sonja PETROVIC, Romain VALABREGUE, Hugo BONIFACE, Rahul GAURAV, François SUZANNE, François-Xavier LEJEUNE, Sana REBBAH, Aurélie KAS, Marie-Odile HABERT, Nadya PYATIGORSKAYA
14:00 - 15:30
#47375 - PG381 Gray Matter Volume Estimation from DWI Segmentation in Ischemic Stroke: A Practical Workflow in a Low-Resource Clinical Setting.
PG381 Gray Matter Volume Estimation from DWI Segmentation in Ischemic Stroke: A Practical Workflow in a Low-Resource Clinical Setting.
Stroke remains a major contributor to global disability and death, ranking fourth in disability-adjusted life-years (DALYs) according to the Global Burden of Disease Study 2021 [1]. In Indonesia, the 2023 National Health Survey reported a stroke prevalence of 0.83% among individuals aged ≥15 years, reflecting a persistent public health burden despite a slight decline from previous years [2,3].
Ischemic stroke, caused by focal infarctions in cerebral, retinal, or spinal regions [4], can alter gray matter volume (GMV). GMV changes have been observed in patients with ischemic thalamic stroke [5] and with atrial fibrillation [6]. Diffusion-Weighted Imaging (DWI), a magnetic resonance imaging sequence sensitive to water diffusion [7], enables early infarction detection [8] and reveals GMV remodeling post-stroke [9]. DWI also enhances gray matter visualization due to its higher diffusion coefficient [7,10].
Given DWI’s accessibility in both tertiary and secondary care settings, this study aims to assess GMV alterations in ischemic stroke patients using a pragmatic, low-resource image processing workflow, with minimal computational dependency.
This study was conducted at the Radiology Department of Syaiful Anwar General Hospital, Malang, Indonesia, using 1.5 T and 3 T MRI scanners, RadiAnt DICOM Viewer, and MATLAB 2014b. The materials included ten axial DWI images from ischemic stroke patients and ten from healthy individuals. As shown in Figure 1, DICOM images were randomly selected and converted to JPEG. Parameters such as TR, TE, and b-values were analyzed. Image processing steps (Figure 2) included RGB-to-grayscale conversion, high-pass filtering, histogram matching, and median filtering to enhance contrast. Segmentation was performed to identify CSF, white matter, and gray matter using thresholding. The segmented image was converted to HSV to facilitate gray matter extraction through masking. Gray matter volume was then calculated and compared between stroke and normal groups to assess structural differences. DWI acquisition revealed long TR values (2,700–5,900 ms), short TE (82–87 ms), and high b-values (1,000 s/mm²), suitable for tissue contrast and diffusion sensitivity. CSF appeared hypointense, while gray matter displayed hyperintensity due to its restricted diffusion. These imaging features were consistent across all patient and control datasets.
Post-segmentation threshold values did not differ significantly between groups (p > 0.05). Mean GMV in ischemic stroke patients was 14.340 cm³, compared to 14.870 cm³ in healthy controls. Although stroke patients consistently showed reduced GMV across age groups, the difference was not statistically significant (p > 0.05).
Variability in pixel spacing, slice thickness, and patient age likely contributed to the absence of significance. However, the segmentation pipeline successfully differentiated gray matter from other tissues in all samples, demonstrating reliability in a basic thresholding framework. This study demonstrates a cost-effective DWI-based image processing workflow capable of extracting and quantifying gray matter volume in ischemic stroke patients. Although limited by a small sample size and the use of basic threshold-based segmentation, the approach remains viable in resource-limited hospital environments where advanced image processing infrastructure is unavailable. The imaging parameters used—long TR, short TE, and high b-values—align with established DWI protocols for optimal infarct visualization and diffusion sensitivity.
While the non-significant GMV reduction may reflect infarction-related atrophy, such changes might be masked by demographic variability and differences in image resolution. Moreover, the lack of statistically significant findings should not diminish the practical importance of this pipeline, especially in developing settings. Importantly, this study highlights the feasibility of GMV estimation without reliance on atlas-based segmentation or machine learning algorithms, offering a starting point for further innovation. Future directions include expanding the sample size, standardizing voxel geometry, and integrating automated segmentation via convolutional neural networks to enhance diagnostic throughput. Using DWI and a straightforward image processing method, this study demonstrates the potential to quantify gray matter volume in ischemic stroke patients, even in environments with limited computational resources. While no statistically significant difference in GMV was observed between stroke and control groups, the methodology reliably segmented gray matter and captured consistent volumetric trends associated with ischemic insult. These results reinforce the value of DWI as a versatile imaging modality for both diagnostic and research purposes. The developed workflow may serve as an initial framework for low-cost, quantitative stroke imaging and can be enhanced with automated tools in future studies for broader clinical use.
Dian Yuliani ALAM (Heppenheim (Bergstraße), Germany), Johan Andoyo Effendi NOOR, Yuyun Yueniwati PRABOWOWATI WADJIB
14:00 - 15:30
#47368 - PG382 Resveratrol protection against cardiac remodeling and ischemia-reperfusion injury in a diet induced prediabetic female rat model: in vivo and ex vivo longitudinal study.
PG382 Resveratrol protection against cardiac remodeling and ischemia-reperfusion injury in a diet induced prediabetic female rat model: in vivo and ex vivo longitudinal study.
Cardiovascular diseases (CVD) are one of the leading causes of mortality and morbidity in patients with type 2 diabetes [1]. Those CVD, such as coronary artery disease or stroke [2–4], can arise before diabetes onset, at the prediabetic stage, with a greater risk for women [5]. Despite being an interesting therapeutic window due to its reversibility [6], first line treatments for prediabetes (lifestyle interventions) do not usually focus on the early cardiac alterations associated with prediabetes [7] and lack cardioprotective effects [8]. Investigating new sex dependent therapeutic strategies is then essential to limit cardiovascular complications in prediabetic women. Resveratrol (RSV) supplementation has previously shown cardioprotective effects on type 2 diabetic female rats [9]. We therefore aimed to compare its effects on the heart of prediabetic female rats to those induced by diet intervention, one of the conventional first line treatments.
40 female Wistar rats were divided into 4 groups fed for 5 months with a standard diet (CTRL), High-Fat-High-Sucrose (HFS) diet, HFS diet supplemented with RSV (1mg/kg/day in drinking water) during the last 2 months (RSV) or HFS diet for 3 months followed by 2 months of standard diet (RSD). We performed a longitudinal in vivo study of cardiac morphology, function and perfusion by magnetic resonance imaging at the 3rd and 5th months. Rats underwent an intraperitoneal glucose tolerance test to evaluate their prediabetic status. Finally, ex vivo experiments on isolated perfused hearts at 5 months were performed to simultaneously study cardiac function (Rate Pressure Product (RPP)) and energy metabolism (Phosphocreatine (PCr), ATP) with 31P magnetic resonance spectroscopy, during an ischemia-reperfusion injury (IR). Experimental protocol is shown in Figure 1. After 3 months of HFS diet, increased myocardial wall thickness (MwT) and perfusion (p<0.01 vs CTRL) were found. HFS diet also induced elevated left ventricular end diastolic volume ((LVEDV) p<0.01 vs CTRL) and mass (LVM) (p<0.05 vs CTRL), along with glucose intolerance at the 5th month (p<0.05). HFS diet increased heart weight to tibia length ratio (HTLR) (p<0.01 vs CTRL), a known cardiac hypertrophy estimator [10]. HFS diet consumption was also associated with alteration of basal myocardial function and tolerance to IR, as shown by impaired RPP (p<0.001) and lower PCr and ATP during reperfusion (p<0.05, p<0.001 vs CTRL). Furthermore, HFS diet induced elevated end diastolic pressure (EDP) throughout the IR protocol (p<0.05 vs CTRL). RSV supplementation restored LVEDV, MwT and LVM (p<0.05 vs HFS), improved glucose tolerance (p<0.05 vs HFS) but exhibited no significant effect on myocardial perfusion. In addition, RSV restored HTLR to CTRL level (p<0.001 vs HFS) and improved myocardial tolerance to IR, characterized by higher RPP (p<0.05 vs HFS) with increased ATP and PCr during reperfusion (p<0.05 vs HFS). Interestingly, RSV also restored EDP over the course of the IR protocol duration (p<0.05 vs HFS). Alternatively, RSD exhibited no effect on LVEDV, MwT, LVM and perfusion impairments (p<0.01 vs CTRL) but restored glucose tolerance (p<0.001 vs HFS) at the 5th month. Furthermore, RSD failed to improve the elevated HTLR and EDP for the whole duration of the IR protocol (p<0.05 vs CTRL) but ameliorated tolerance to IR, characterized by increased RPP (p<0.05 vs HFS) and higher PCr and ATP during reperfusion (p<0.001, p<0.01 vs HFS). 5 months of HFS diet induced prediabetes, deleterious cardiac remodeling, increased myocardial perfusion but also reduced tolerance to IR injury, characterized by decreased myocardial function and PCr, ATP, as previously found in other prediabetic models [11–13] but also patients [14–16]. The two therapeutic approaches exhibited different cardioprotective effects here. RSV protected against deleterious cardiac remodeling induced by HFS diet in vivo and ex vivo but also improved glucose tolerance and myocardial susceptibility to IR injury. Interestingly, 2 months of a return to standard diet had no effect on deleterious cardiac remodeling induced by HFS feeding. However, RSD restored glucose tolerance to control levels but only improved myocardial susceptibility to IR injury. This observation could be explained in part by the lack of an effect on deleterious cardiac remodeling. Interestingly, RSV, despite maintained HFS feeding, exhibited a stronger cardioprotective effect than 2 months of return to standard diet. Further studies are warranted to uncover the mechanisms involved, with a particular interest in mitochondrial function and oxidative stress. However, these results suggest that RSV supplementation could be an interesting approach to address the cardiac alterations associated with prediabetes, especially for women, which are at greater risk of CVD than men.
Alexis JOUENNE (MARSEILLE 05), Isabelle VARLET, Christophe VILMEN, Frank KOBER, Monique BERNARD, Martine DESROIS
14:00 - 15:30
#47853 - PG383 Evaluating the impact of pregnancy on cerebrovascular function.
PG383 Evaluating the impact of pregnancy on cerebrovascular function.
Cerebrovascular reactivity (CVR), the brain's ability to regulate blood flow in response to stimuli, is a critical indicator of cerebrovascular health. Impaired CVR has been shown in neurodegenerative diseases, such as Alzheimer’s [1], which has disproportionally higher prevalence in women [2]. Pregnancy represents a female-specific life event that leads to significant neuroanatomical changes, including in grey matter (GM), that remain years after baby delivery [3]. Women with a history of hypertension during pregnancy have been shown to exhibit reduced CVR [4] and increased dementia risk [5] compared to women with normotensive pregnancies, however the impact of pregnancy without hypertension on cerebrovascular health is not clear. In this work we investigate whether pregnancy history is associated with changes in global and regional CVR.
Multiple post labelling delay (multi-PLD) pCASL data (6 PLDs 250-1500ms, labelling duration 1400ms, 3.5x3.5x4.5mm³) from 40 female subjects was acquired on a 3T Siemens Prisma scanner during two conditions, normocapnia (6 min) and hypercapnia (5% CO2, 3 min). Six participants were excluded due to data quality issues. Analysis included two groups of women post-partum (14.9±5.8 months post-delivery): (1) normotensive pregnancy (n=13, 34.5±3.3 years), (2) hypertensive pregnancy (n=3, 33.3±2.1 years), and an age-matched group with 18 women that have never been pregnant (31.7±5.1 years).
ASL data were analysed with FSL tools, including BASIL [6], yielding maps of CBF and arterial arrival time (AAT). Structural images were segmented to retrieve GM masks. Internal carotid artery velocities from phase contrast data were used to estimate labelling efficiency [7] and perfusion maps calibrated by hemisphere- and subject-specific labelling efficiency. CVR was calculated as the absolute change in perfusion over the change in end-tidal pCO2. Global and GM CVR and CBF metrics were compared using t-tests (p<0.05). A voxelwise analysis of CVR and resting CBF was performed using Randomise (threshold-free cluster enhancement, 5000 permutations) with age as covariate. No statistically significant differences were found in either global or GM CVR between women with no history of pregnancy and women with a history of normotensive pregnancy (Figure 1). Similarly, no differences were detected in normo- or hypercapnic CBF between these groups (Figure 2). Voxelwise analysis did not identify any regions of statistical significance. Descriptively, CVR metrics were lower in the group of women with hypertensive pregnancy disorders (Figure 3), but statistical analysis was precluded by the limited sample size. In contrast with previous reports of brain anatomical changes that persist years after baby delivery, cerebrovascular changes, evaluated through CBF and CVR, seem to be preserved following normotensive pregnancy. The preliminary observation of reduced CVR in women with history of hypertensive pregnancy disorders aligns with previous research and calls for further investigation in a larger cohort. It also highlights how CVR, as a functional measure, complements measures of CBF in assessing cerebrovascular function. Using ASL MRI with hypercapnic challenge, we found no significant differences in global CBF and CVR between postpartum women with normotensive pregnancy history and women that have never been pregnant, suggesting preserved cerebrovascular function despite other research finding structural brain adaptations. These findings contribute to the growing body of research on the impact of sex-specific life events on brain health.
Lise KLAKSVIK (Oxford, United Kingdom), Daniel BULTE, Joana PINTO
14:00 - 15:30
#47741 - PG384 MRI-based comparison of aortic blood flow in rabbits under physiological and extracorporeal circulation.
PG384 MRI-based comparison of aortic blood flow in rabbits under physiological and extracorporeal circulation.
Extracorporeal circulation (ECC) is vital in cardiac surgery [1], yet its effects on flow dynamics and tissue perfusion are still debated. Comparing ECC with physiological blood flow is essential for understanding these effects. MR imaging of moving organs requires advanced tracking for reliable results [2]. In addition, the wide range of flow velocities during ECC present specific challenges for phase-contrast MRI.
To explore the impact of the different perfusion types, we compared aortic flow during ECC with physiological conditions in a rabbit model using MRI. Non-pulsatile flow was measured using an MR-compatible ECC set up [3], while physiological flow was assessed using navigator-guided, retrospectively gated imaging, both in anesthetized rabbits.
All MR data were acquired from healthy rabbits using a 9.4 T Bruker BioSpec equipped with a Tx/Rx birdcage coil. Rabbits were examined under 3 perfusion conditions —physiological, antegrade, and retrograde—each tested on 7 rabbits. Flow MRI covered the entire aorta and major branching vessels, divided into thoracic, abdominal, and pelvic segments, achieving an isotropic spatial resolution of 0.5 mm.
For the continuous ECC blood flow, a specialized MRI protocol utilized 3 Venc values (200, 50, and 20cm/s, TR/TE: 7.5/3.5ms) allowing for accurate reconstruction of a wide velocity range (Fig. 1). For physiological blood flow, Bruker’s FLOWMAP was adapted by adding a navigator, eliminating the need for complex physiological recording (TE/TR: 4/9.3ms, Venc: 100cm/s, 14 oversamplings), and retrospective gating was used to reconstruct pseudo-dynamic physiological flow (Fig. 2) [4]. All datasets were processed using ROMEO phase-unwrapping [5] and polynomial surface fitting on static tissue for background phase correction. Both types of perfusion during ECC were conducted using similar volumetric inflow rates, with only minor variations between individual measurements.
In the ascending aorta, significant differences in volume flow rates were noted between antegrade and retrograde perfusion, while antegrade perfusion and physiological perfusion yielded comparable results. In the biologically downstream segments of the aorta, including thoracic and abdominal areas, substantial differences in blood volume flow rates were observed among all three types of perfusion, with antegrade perfusion showing the highest volume flow rates. In the descending aorta, the volume flow rates for antegrade perfusion were three times higher than those for retrograde perfusion, while the value for physiological flow was intermediate between the two (Fig. 3). In the distal aorta, volume flow rates were similar for physiological and antegrade perfusion, whereas retrograde perfusion exhibited significantly higher rates. Conversely, in the supraaortic and visceral branches, no significant differences were found among the three types of perfusion. Since EEC perfusion aims to replace cardiac function, it is preferable to achieve volume flow rates that are comparable to physiological conditions, even though pulsatility is absent. Evaluating perfusion conditions specific to the EEC scenario and comparing them to physiological conditions can provide valuable insights for developing optimal procedures for human applications.
Using the triple VENC technique employed here, it was possible to accurately quantify different flow velocities, particularly in the inflow region of the cannula as well as in peripheral areas. As expected, due to the varying flow directions, differences in flow rates between antegrade and retrograde perfusion could be quantified. Interestingly, rather than following a strict pattern, differences between the two EEC methods and the physiological condition varied across different vascular regions.
A significant challenge encountered was the segmentation of small peripheral vessels. For future studies, higher spatial resolution will be necessary to address this limitation. Using a dedicated triple Venc approach for ECC and a navigator-based method for pseudo-dynamic physiological blood flow, different perfusion strategies could be systematically compared in a rabbit model. The presented methodology enables reliable evaluation of different ECC approaches in vivo, providing a robust foundation for future testing and optimization of new perfusion strategies intended for clinical application in humans.
Jonah SCHRAUDER (Göttingen, Germany), Anna Kathrin ASSMANN, Alexander ASSMANN, Tor Rasmus MEMHAVE, Amir MOUSSAVI, Susann BORETIUS
14:00 - 15:30
#47732 - PG385 Influence of hormonal contraception on cerebral perfusion.
PG385 Influence of hormonal contraception on cerebral perfusion.
Arterial Spin Labeling (ASL) is a non-invasive MRI technique widely used to measure cerebral perfusion. However, its application is often limited by substantial intra- and inter-subject variability, which complicates the differentiation between physiological and pathological changes [1]. Despite increasing interest in the female brain, the effects of hormonal contraception on cerebral perfusion and hemodynamics remain poorly understood. This study aims to address this gap by investigating the impact of hormonal contraception on cerebral perfusion (CBF) and arterial transit times (ATT) using ASL MR imaging.
MRI data in 9 women with a natural menstrual cycle (NC) and 15 women using hormonal contraception (HA, Desorelle 20) were obtained on a Siemens 3T Prisma Fit MRI scanner (64 channel head coil, UGent core facility Ghent Institute for functional and Metabolic Imaging (GIfMI)). Data were acquired at three (for NC group) and two (for HA group) timepoints during the menstrual cycle, for three cycles, resulting in nine datasets per volunteers. ASL was acquired using a multi-time point pulsed ASL (TR = 3500ms, labeling duration = 1.8s; post-labeling delay’s from 0.3s to 3.1s, increments 0.3s, 1 label-control pair), and processed using the Bayesian Inference for Arterial Labeling (BASIL) toolset [2] from FSL (distortion correction with field map, model with macrovascular component and spatial priors, voxelwise calibration). Regional CBF and ATT were quantified for nine brain regions based on the MNI152 atlas (caudate, cerebellum, frontal lobe, insula, occipital lobe, parietal lobe, putamen, temporal lobe, and thalamus). Additionally, blood samples were drawn to analyze concentration of sex hormones (estradiol (E2), progesterone (Prog), follicle stimulating (FSH), and luteinizing hormone (LH)). rCBF, ATT and sex hormones of both groups were compared using a Mann-Whitney test with a False Discovery Rate correction and the significance level of alpha = 0.05. A significant lower level of LH (mean difference = -4.45 U/L, p<0.001), FSH (mean difference = -2.25 U/L, p-value < 0.05), E2 (mean difference = -45.40 ng/L, p-value < 0.001) and Prog (mean difference = -0.25 μg/L, p<0.001) was measured in the women using contraception compared to the women wih a natural menstrual cycle. Significantly higher perfusion in HA compared to NC was measured in all regions except the cerebellum, with differences ranging from 1.25 ml/100g/min (temporal lobe) to 14.09 ml/100g/min (thalamus) (Figure 1). Additionally, in HA, compared to NC, a significant higher ATT was measured in the cerebellum (mean difference = 0.031s, p<0.001), and lower in the frontal (mean difference = -0.019s, p<0.01) and parietal lobe (mean difference = -0.014s, p<0.05) and the thalamus (mean difference = -0.035 ml/100g/min, p<0.001)(Figure 2). No statistical significant differences in ATT were found in the other regions. This study highlights the impact of hormonal contraception on cerebral perfusion and arterial transit time. HA users showed higher CBF in most brain regions, higher ATT in the cerebellum and lower in the frontal, parietal lobes, and thalamus. These findings emphasize the need to consider hormonal status in neuroimaging studies, as HA can influence CBF and ATT. Limitations include a small sample size, and future research should explore larger cohorts and various contraceptives. Hormonal contraception use significantly impacts cerebral perfusion and ATT measurements, highlighting the need to account for hormonal status in perfusion imaging studies. These results contribute to a better understanding ofthe influence of sex hormones on cerebral hemodynamics.
Soetkin BEUN (Ghent, Belgium), Ceulemans JULIE, Thomas OKELL, Eric ACHTEN, Joana PINTO, Patricia CLEMENT
14:00 - 15:30
#47802 - PG386 Enhanced hand angiography without contrast agents: One-slab, PPU-triggered QISS with adapted echo times.
PG386 Enhanced hand angiography without contrast agents: One-slab, PPU-triggered QISS with adapted echo times.
Since its introduction about 15 years ago, Quiescent-Interval Single-Shot (QISS)1,2,3 has been widely used for peripheral MR angiography, e.g. for peripheral arterial disease (PAD)2,3.
In a recent publication4, QISS was used to acquire high resolution hand angiograms using ECG triggering at 1.5 T. However, strenuous pre-scan preparation steps, prominent background signal and the presence of artefacts such as discontinuity and susceptibility were reported.
Here, we ventured to address these issues by using a one-slab excitation, Peripheral Pulse Unit (PPU) out-of-phase5 imaging on a latest generation, high performance 3T system.
Healthy volunteers were scanned in prone position with one hand extended above the head (“superman” position) using a product 2D QISS sequence at 3 T with one or several “slab” adjustment volumes (120° slice selective excitation, Cartesian True-FISP (Trufi) readout, venous saturation band = 70 mm; MAGNETOM Cima.X, max. gradients / slew rate 200 mT/m(s), 16-ch. Hand-wrist coil, Siemens Healthineers).
Two echo times, TE = 2.42 ms (in phase), and TE = 3.5 ms (out-of-phase) were chosen by setting the receiver bandwidth (BW) to 651 Hz/Px (protocol 1), 347 Hz/Px (protocol 2, full echo) and 195 Hz/Px (protocol 3, 2/3 partial echo), respectively. Note that the sequence didn’t allow for setting the TE separately yet.
Other parameters were: Quiscent-interval TI = 345 ms, Peripheral Pulse Unit (PPU) triggering, Single 3D slab reconstruction, TR = 687.61ms, interpolated voxel size = 0.2x0.2x1.0 mm3, matrix size= 640X416p, 256 slices, FOV = 165x108 mm2. Contrast to Noise Ratio (CNR) was estimated in three arteries: Ulnar, Radial, Proper Palmar Digital arteries in three distinct imaging slices using:
CNR=(S1−S2)/σ
S1 is the mean signal of a region of interest (ROI) in an artery in a single slice, S2 is the mean signal of a ROI of background tissue BT, σ is the standard deviation of the noise outside the object. Using a single slab averted all block-like artefacts reported before (Fig. 1), 4 and resulted in good quality arterial QISS angiograms in short scan times of 3:36 min (Fig. 2).
Using the PPU instead of the ECG for triggering did not deteriorate the apparent image quality; the arteries of the hand were well visualized with no additional artefacts.
Using the same out-of-phase-TE, but increasing the bandwidth, reduced the tissue signal by signal (BT, Protocol 3) / signal (BT, Protocol 2) = 1.2 (Fig. 4). Of course, these results are affected by the echo time and bandwidth – see discussion.
For all protocols, the maximum intensity projections (MIP) of the data exhibited similar arterial structures (Fig. 2). Susceptibility artefacts manifested as smearing of the arteries more prominently at lower bandwidths, discontinuities were present in one location at BW = 195 (Fig 2c).
The CNR has increased for smaller bandwidths and for out-of-phase TE across all three arteries (Fig. 3), except for the radial artery in volunteer 2 (in color red Fig. 3). Using a single instead of multiple adjustment slabs resulted in MIPs free of the block artefact (Fig. 1), likely because more uniform adjustment parameters were used.
PPU has provided good results for hand angiography, although previous studies1,2,4 deemed PPU to be inferior. The difference may be caused by faster PPU electronics of the system used.
Setting TE to cause fat and water spins to be out-of-phase is an established method to reduce lipid signals (TE = 1.23 ms, 3.69 ms, 6,15 ms, 8.61 ms, …. at 3T). When we set TE close to the out-of-phase condition, the background tissue around the arteries is noticeably darker, indicating better suppression of the static tissue, and consequently, enhancing the subjectively observable contrast between the arteries and the static tissue (Fig.1b and 1c, CNR in Fig. 2). For all three arteries, CNR was higher for TE= 3.4 ms/ BW = 195Hz/Px.
This imaging protocol have yielded a near full visualization of the major hand arteries without requiring the lengthy pre-scan preparation set-up procedures (moisturizing hands, using clay mold)4.
Of course, the receiver bandwidth has an effect on CNR, too. Higher bandwidths are associated with less signal and higher noise due to the higher sampling rate. This fact skews the comparison of CNR in favor of data with lower bandwidths (Fig. 3). On the other hand, lower bandwidths risk exacerbating susceptibility artifacts (Fig. 2). We are currently implementing a sequence that allows for setting TE individually. A high bandwidth, out-of-phase TE sequence appears to be ideal to minimize artifacts and maximize static tissue suppression. The developed protocols, relying on one-slab-adjustments, PPU triggering and out-of-phase imaging, provided high quality angiograms of the human hand without contrast agents, and averted the issues reported before. More experiments are needed to isolate the individual effects.
Mayar FAHED (Kiel, Germany), Mona SALEHI RAVESH, Mariya PRAVDIVTSEVA, Monika HUHNDORF, Lynn Johann FROHWEIN, Robert R. EDELMAN, Marcus BOTH, Olav JANSEN, Jan-Bernd HÖVENER
14:00 - 15:30
#47686 - PG387 Effect of catheter ablation of atrial fibrillation on the renal blood flow measured by phase-contrast MRI - A pilot study.
PG387 Effect of catheter ablation of atrial fibrillation on the renal blood flow measured by phase-contrast MRI - A pilot study.
It is known that atrial fibrillation (AF) and chronic kidney disease (CKD) are common conditions which are closely related and usually coexist. Each disease influences progression of the other. However, it is not well understood yet how AF affects renal blood flow. 2D phase-contrast magnetic resonance imaging (2D PC MRI) is a non-invasive method capable providing the functional information about renal artery blood flow (RABF) and quantify it. Therefore, we utilized 2D PC MRI to compare RABF in the same patient during AF and consequently in sinus rhythm (SR) after catheter ablation (CA) in general anaesthesia.
This pilot study enrolled 7 patients (age: 62 ± 13 years, F/M: 2/5) who underwent CA for AF in general anaesthesia. All subjects provided written informed consent with the participation in the study. The study was conducted in compliance with the principles of the Declaration of Helsinki and with the approval of local ethics committee.
All the patients underwent MR examination 24 hours before CA and consequently 24 hours after CA. The examination was performed in the supine position during breath-hold exhalation using 3T VIDA MR system (Siemens Healthineers, Germany) equipped with 30-channel surface coil and 32-channel spine coil. MRI protocol included T2 TruFI sequences in 3 orthogonal orientations (thickness: 3 mm) for renal anatomy visualization and 2 sets of 2D PC MR sequences (1. FOV 250x250 mm, flow-encoded velocity range (VENC) = 100 cm/s, TR/TE = 40.80/2.82 ms; 2. FOV 200x200 mm, VENC = 80 cm/s, TR/TE = 40.64/2.99 ms; NA = 1, flip angle = 20°). The flow data were obtained in a plane perpendicular to the right and left renal artery (RA), approximately 10 mm from the ostium. 2D PC MR measurements were repeated 2 or 3 times to precise RABF quantification. Retrospective ECG or pulse triggering with RR-based arrhythmia rejection was used.
All the data sets were normally distributed. Paired t-tests were used for statistic evaluation of the changes in patients before and after CA. P-value < 0.05 was considered statistically significant. All patients presented with normal SR after CA. No significant differences in renal function parameters were found between AF and SR (creatinine before CA: 94±22 µmol/l, after CA: 97±25 µmol/l; cystatin C before CA: 1.1±0.3 mg/l, after CA: 1.1±0.3 mg/l). Paired t-test showed significantly decreased cross-section area (ROIs) of both renal arteries, increased RABF and increased renal blood peak velocity in each patient with SR after CA compared to the state in AF before CA (Figure 1, Table 1). 2D MR PC measurements were repeated 2 or 3 times for each FOV and VENC assessment to precise selection of RA cross-section area. Considering the size of the RA caliber and the realistically achievable spatial resolution when measuring RABF, it was not possible to reliably use automatic ROI delineation. RA ROIs needed to be selected manually which increases the probability of bias selection. The relative error for all evaluated parameters ranged from 10 – 20 %. Although the difference in RA cross-section areas before and after CA may have been biased by both ROIs selection error and renal artery wall pulsatility, neither RABF nor renal blood peak velocity can be influenced by this difference. Restoration of SR following CA of AF is associated with an increased renal blood peak velocity and higher renal blood flow. These preliminary results require further investigation and verification in future studies.
Acknowledgements
This study was supported by the project National Institute for Research of Metabolic and Cardiovascular Diseases (Programme EXCELES, Project No. LX22NPO5104) – Funded by the European Union – Next Generation EU. This work was also funded by the project (Ministry of Health, Czech Republic) for development of research organization 00023001 (IKEM, Prague, Czech Republic) – Institutional support.
Dita PAJUELO (Prague, Czech Republic), Predrag STOJADINOVIĆ, Monika DEZORTOVÁ, Milan HÁJEK, Josef KAUTZNER, Jaroslav TINTĚRA
14:00 - 15:30
#46604 - PG388 Characterising high-risk carotid plaques and endothelial (dys)function using non-contrast enhanced MRI.
PG388 Characterising high-risk carotid plaques and endothelial (dys)function using non-contrast enhanced MRI.
Carotid atherosclerosis remains a major risk factor for stroke in western countries[1]. While clinical management typically depends on stenosis severity, stenosis alone often fails to predict stroke risk accurately [2,3]. Treatment decision is currently based on the severity of stenosis, even though, clinical trials have shown that stenosis alone may not reliably predict cardiovascular risk in individual patients. Plaque composition and compromised endothelial function (in response to vasodilating stressors) are crucial in plaque development and the occurrence of cardiovascular events[4–6]. Consequently, an imaging method to detect features of high-risk plaque and luminal stenosis simultaneously would have added value in patient diagnosis. The recently proposed BOOST (Bright- and Black-blOOd phase-SensiTive) sequence offers simultaneous, co-registered, contrast-free bright- and black-blood imaging in a single scan, enabling both lumen and vessel wall visualisation[7]. BOOST has been previously used at 1.5T to identify high-risk coronary plaque features (thrombus and intraplaque haemorrhage)[8,9] and for anatomical assessment of the aorta[10]. However, the application of BOOST at 3T for 3D high-resolution imaging of carotid plaques has not been explored. Here, we optimised the iT2prep-BOOST sequence for carotid vessel wall imaging at a 3T scanner. This study was designed to selectively identify patients with high-risk carotid atherosclerosis by detecting compositional features of high-risk plaques and assessing endothelial (dys)function in a fast, high-resolution, contrast-free MRI session.
Building on our previous work [7], we have implemented an accelerated, 3D free-breathing, motion-corrected iT2 prep-BOOST research sequence on a 3T MR system (MAGNETOM Vida, Siemens Healthineers AG, Forchheim, Germany) for carotid artery imaging (Fig. 1). iT2 prep-BOOST allows for simultaneous bright and dark-blood imaging Images were acquired using a 64-channel head and neck matrix coil (Head/Neck 64, Siemens Healthcare) in 4 healthy subjects (female; mean age 34±14) with cardiac synchronisation via peripheral pulse sensor unit to optimise the acquisition parameters (data not shown). Patients undergoing carotid endarterectomy (CEA) (n=6) were recruited (Fig.2A). The iT2 prep-BOOST sequence was used for carotid plaque imaging at high resolution (0.9 x 0.9mm) and speed (~6 min). Endothelial (dys)function was measured using a phase-contrast MRI protocol before and 120 seconds after a cold-pressor test (CPT). Plaque-to-muscle ratio (PMR=SI plaque/SI reference muscle) using the black-blood images and changes in vessel area and blood flow were analysed. Tissue was collected postoperatively to validate the imaging data. Results of the proposed simultaneous lumen and vessel wall BOOST sequence are shown for three patients. MR angiography and examples of bright and black-blood images acquired with iT2 prep-BOOST, show excellent delineation of the carotid artery lumen and vessel wall in all patients (Fig. 2B). The PMR measured on MRI was higher in plaques presenting with intraplaque haemorrhage as verified by en face tissue inspection (Fig. 2C). Assessment of endothelial vasomotor response to CPT showed vasocontraction in all three cases (negative % change in area) and reduction or no change in blood flow (negative or close to zero). This study is the first to demonstrate the feasibility of a rapid, high-resolution MRI approach for simultaneous assessment of high-risk features in carotid plaques and endothelial function under 20 minutes. The iT2-prep-BOOST sequence provided clear delineation of the arterial lumen and vessel wall, and showed a higher plaque-to-muscle ratio in high-risk plaques with intraplaque haemorrhage. Cold pressor testing showed endothelial dysfunction characterised by no changes or reduced vessel area and blood flow at sites of plaque. This integrated approach offers a time-efficient method enabling the assessment of both plaque composition and endothelial cell function in a single MRI session, with the potential to improve risk stratification of carotid atherosclerosis. In this translational study, we demonstrate for the first time that in vivo imaging of both high-risk plaque features and endothelial (dys)function can be assessed in a single MRI session at high resolution and in under 20 minutes. Such a strategy that provides both anatomical and functional assessment of the plaque may enable better stratification of atherosclerosis and selection of patients at risk.
Nadia CHAHER (London, United Kingdom), Darshan BAKRI, Karl P KUNZE, Ivan KOKHANOVSKYI, Claudia PRIETO, René M BOTNAR, Prakash SAHA, Alkystis PHINIKARIDOU
14:00 - 15:30
#46705 - PG389 Predictors of Cerebral Microbleeds in CADASIL: Two analytical approaches.
PG389 Predictors of Cerebral Microbleeds in CADASIL: Two analytical approaches.
Cerebral Autosomal Dominant Arteriopathy with Subcortical Infarcts and Leukoencephalopathy (CADASIL) is the most frequent hereditary cerebral small vessel disease (SVD) worldwide. The condition caused by characteristic mutations of the NOTCH3 gene leads to recurrent stroke, motor impairment, gait disturbances and cognitive decline [1]. Common magnetic resonance imaging (MRI) findings include white matter hyperintensities on T2-weighted images, lacunes and brain atrophy. Cerebral microbleeds (CMBs) described as small hypointense areas related to focal iron deposits on MRI sequences are observed in about one third of patients [2]. The predictors of CMBs in CADASIL are unclear, as the exact cause of their emergence and growing number remain poorly understood. Analyzing CMBs is however challenging for two main reasons: 1) CMB count represents a discrete and typically zero-inflated variable with a long-tailed distribution [3]; 2) CMB counting becomes less reliable when their number increases (see colleague submitted abstract [4]). In the present study, we used a quantitative and semi-quantitative approach to assess CMBs in CADASIL and sought to compare differences between CMB predictors obtained using two different analytical strategies.
We applied a two-step modeling framework to data obtained from 517 CADASIL patients recruited at the National Referral Center in France. Key variables were considered in addition to CMBs for analysis such as age, sex, vascular risk factors, the NOTCH3 mutation location, and key biological and neuroimaging information. In the first approach, the presence of CMBs was modeled using binary logistic regression (Logit) and thereafter CMB count (≥1) using a truncated negative binomial regression (NB). In the second approach, a binary logistic regression was also used at first, and thereafter an ordinal logistic regression (OLogit) based on four number categories. Continuous variables were standardized. Missing data were handled via mean imputation (imaging results) or via multiple imputation by chained equations (other data). Covariates were first screened for multicollinearity. A univariate analysis was then used to exclude those unlikely related to CMBs (p > 0.3). Age, hypertension, other neuroimaging measures and mutation location were retained in all models. The final model predictors were selected using stepAIC (OLogit) or LASSO (Logit and NB). When possible, model performance was assessed using a 70/30 train-test split. Model fit was evaluated with log-likelihood, Akaike Information Criterion (AIC), and Bayesian Information Criterion (BIC), while predictive performance was assessed using confusion matrices and mean absolute error (MAE). All analyses were conducted in R Statistical Software version 4.4.3 [5]. For both approaches, the Logit model identified age, hypertension, the number of lacunes, and diastolic blood pressure as predictors of CMB occurrence (Table 1). Using quantitative data, the NB model showed that brain parenchymal fraction, number of lacunes, MRI sequence, hemoglobin level and both systolic and diastolic blood pressure predicted the number of CMBs (Table 2). In contrast, when CMB were considered by number categories, the OLogit model identified the number of lacunes as the unique predictor (Table 3). In terms of predictive performance, the Logit model demonstrated strong classification ability, with high sensitivity and a robust F1 score, though it tended to overpredict the presence of CMB. The NB model exhibited moderate predictive accuracy, underestimating higher CMB counts, likely due to challenges in modeling overdispersion. The OLogit model showed limited capability in correctly classifying higher CMB categories. Our two analytical approaches yield different sets of CMBs predictors in CADASIL. The NB and OLogit models are not comparable. Choosing a quantitative or semi-quantitative measures of CMBs leading to different analytical procedures appears thus crucial and should align with the exact underlying biological question. If the goal is to identify all predictors of CMBs, the first approach should be adopted despite the lack of precision in higher counts. If the goal is to obtain an accurate prediction, the second approach is preferable at the cost of losing information. Limitations of this study include the moderate sample size and CMB detection measures. Additional analysis of longitudinal data should help improve predicting CMB in CADASIL. The results of this study further illustrate that the decision to quantify a biomarker such as CMBs should be based primarily on the ultimate objectives sought. Analysis of such data leads to a trade-off between the desired predictive performance, the complexity of the model to be developed, and the type of sample to be analyzed. In the future, enriching the information with longitudinal MRI data should further improve the prediction of CMBs in CADASIL.
Laura TINTORE CARBONELL (Paris), Jessica LEBENBERG, Louis LAMBERT, Mohamed SAICHI, Hugues CHABRIAT
14:00 - 15:30
#46908 - PG390 A reliability analysis for cerebral microbleeds counting in CADASIL.
PG390 A reliability analysis for cerebral microbleeds counting in CADASIL.
Cerebral Autosomal Dominant Arteriopathy with Subcortical Infarcts and Leukoencephalopathy (CADASIL) is a monogenic cerebral small vessel disease (cSVD) caused by stereotyped cysteine mutations of the NOTCH3 gene encoding a receptor of smooth muscle cells and pericytes of arterioles and capillaries. This condition may lead to stroke, motor impairment, gait disturbances and cognitive decline [1]. On cerebral MRI, several MRI markers are observed during the disease course, including cerebral microbleeds (CMBs). CMBs are small hypointense dots on T2* and SWI sequences related to focal iron deposits (see Fig1). They are observed in 1/3 of patients [2]. Because of the higher sensitivity of SWI, the number of CMBs may be found higher than on T2*. As the individual identification of CMBs may be tedious both visually and quantitatively, some researchers labelled CMBs as ‘certain’ or ‘uncertain’, and others proposed to categorize their number in different classes [3–7]. As the method used to analyze CMBs may largely impact the identification of the potential predictors of such lesions (see companion submitted abstract [8]), we here assessed how the number of CMBs, the method used for counting and the type of MRI sequence can influence the observer agreement in repeated and independent assessments.
Thirty T2* and thirty SWI MRI were acquired respectively on a 1.5T GE scanner or on a 3T SIEMENS machine in CADASIL patients included in the National French referral center for rare vascular diseases of the eyes and brain (https://www.cervco.fr). On each scan, CMBs were visually identified twice by a trained scientist (Rater1 performed Reviews 1 and 2), and once by an expert neurologist (Rater2). Counts were then transformed in CMBs categories according to the CADAMRIT instrument (CADAMRITlike items) [7]. The continuous and categorial agreements were evaluated with the ICC3 and weighted kappa scores respectively [9,10]. A linear regression and a category-based analysis were performed to evaluate the influence of the burden of CMBs onto the results. For each test, the MRI sequence was considered as a factor that could potentially influence the results. In the T2* sub-dataset, 63% of individuals were women, the mean age was 60.22 +/- 9.04 years. In the SWI sub-dataset, 50% of individuals were women, the mean age was 58.85 +/- 7.84 years.
When CMBs were assessed as a continuous variable on T2*, the ICC3 reached 0.95 and 0.87 for the intra- and inter-rater assessments respectively. On SWI, the same parameters reached 0.87 and 0.65 for the intra- and inter-rater evaluations respectively (p always <0.001). Linear regression analysis between the absolute difference of counting and the mean of values, showed that the intra- and inter-rater agreements decreased significantly with the number of CMBs, especially when using the SWI sequence. For the intra-examination study, the R2 was 0.732, and the interaction between the mean value and the MRI sequence was significant with p = 0.032. For the inter-rater study, the R2 was 0.616, and the interaction between the mean value and the MRI sequence was significant with p < 0.0001. Results are detailled in Fig2. The absolute difference between CMBs counting against the mean count are displayed on Fig3.
For the categorical classification of CMBs on T2*, the weighted-Kappa reached 0.97 and 0.74 for the intra- and inter-rater examinations respectively. For CMBs counting on SWI, these scores reached 0.94 and 0.83 for the intra- and inter-studies respectively. All tests were statistically significant (p<0.001). Because of the limited sample size, a category-based analysis could not be evaluated. The heatmaps presented in Fig4 showed a high intra-rater agreement. More discrepancies were found for the inter-rater, especially for T2* data. The results of this study showed that, globally, the intra- and inter-observer agreements of the number of CMBs on T2* and on SWI were always relatively high, both for continuous and categorial countings. They also showed that the agreement for continuous countings decreased significantly with the number of lesions to be detected. As the SWI sequence is very sensitive to iron deposits, such an effect appeared larger for SWI than for T2*images. Because of the limited sample size, the potential effects of evaluation by number categories on inter or intra-rater agreement was not assessed. Our results emphasized that the reliability of CMBs identification mainly depends on the burden of lesions to be detected. Future analysis will investigate whether an automated segmentation may improve the counting reliability [6]. Pending further in-depth studies, we suggest a cautious interpretation of results including high number of CMBs [8].
Jessica LEBENBERG (Paris), Mohamed SAICHI, Laura TINTORE CARBONELL, Louis LAMBERT, Hugues CHABRIAT
14:00 - 15:30
#47735 - PG391 Longitudinal 3D-DCE MRI Evaluation of Placental Perfusion in a Rat Model of Reduced Uterine Perfusion Pressure (RUPP).
PG391 Longitudinal 3D-DCE MRI Evaluation of Placental Perfusion in a Rat Model of Reduced Uterine Perfusion Pressure (RUPP).
The placenta is a dynamic organ essential for supporting fetal growth and development, functioning as the critical interface for maternal-fetal exchange of nutrients, gases, and signaling molecules. Its normal development and function are crucial for a healthy pregnancy, yet can be compromised in conditions such as preeclampsia (PE) and fetal growth restriction (FGR), both of which are major contributors to perinatal morbidity and mortality (1). One commonly used preclinical model to study these conditions is the Reduced Uterine Perfusion Pressure (RUPP) model, which mimics key features of PE by surgically decreasing uterine blood flow (2). In this study, we use longitudinal Dynamic Contrast-Enhanced (DCE) MRI to non-invasively assess changes in placental perfusion across multiple gestational days in rats subjected to RUPP compared to normal pregnancies. This approach enables evaluation of both the acute effects immediately following the surgery and the potential compensatory adaptations that occur later in gestation.
Animal experiment:
Our study used pregnant Sprague Dawley rats, divided into two groups: normal pregnancies (NP) (n=8) and RUPP pregnancies (n=10). The RUPP model was established by clipping the ovarian arteries and abdominal aorta to reduce uterine blood flow.
Each animal underwent three MRI sessions starting at gestational day (GD) 14, immediately after surgery, followed by sessions on GD15 and on either GD16 or GD18. After the final MRI session, animals were sacrificed.
3D-DCE MRI:
To assess perfusion, DCE MRI was conducted using a 7T scanner (MR Solutions, Guildford, UK), utilizing a 3D RF-spoiled gradient-echo sequence with the following parameters: TR/TE = 8/2 ms, flip angle = 25°, FOV = 60 x 60 x 60 mm³, matrix = 128 x 96 x 96. K-space filling was done using a pseudo-radial scheme in the two phase-encoding directions and a tiny golden-angle increment of 16.95 degrees between successive spokes. The pseudo-radial acquisition was repeated for 11 minutes. Subsequently, data was reconstructed with a temporal resolution of 22 seconds using a compressed sensing reconstruction algorithm with a total variation regularization in the temporal domain. Semi-quantitative analysis of the DCE MRI signal intensity-time curves was performed using three perfusion parameters: area under the curve (AUC), peak enhancement, and time to peak (TTP). On GD14, placental perfusion was significantly impaired in the RUPP group compared to the NP group. Specifically, AUC was markedly reduced (RUPP: 52,000 ± 7,200 vs. NP: 70,500 ± 6,800; p < 0.001), peak enhancement values were lower (RUPP: 102 ± 15 vs. NP: 145 ± 10; p < 0.001), and TTP was significantly delayed (RUPP: 288 ± 21 ms vs. NP: 195 ± 18 ms; p < 0.01)
At subsequent time points, perfusion metrics in the RUPP group showed progressive recovery. By GD18, AUC and peak enhancement values in RUPP placentas were comparable to those observed in the NP group (AUC: RUPP: 68,000 ± 8,200 vs. NP: 67,500 ± 7,400; Peak: RUPP: 140 ± 12 vs. NP: 138 ± 11). TTP differences also diminished by GD18. The results demonstrate that placental perfusion was significantly reduced in the RUPP group compared to the normal pregnant group at the earliest post-surgical time point. This reduction was evident across all measured parameters, including AUC, peak enhancement, and TTP, indicating impaired placental blood flow following the surgical intervention.
At later gestational days, the perfusion parameters in the RUPP group showed progressive changes. By the final time point, values for AUC and peak enhancement were similar between the RUPP and control groups, and the initial delay in TTP had largely resolved. This study highlights the dynamic adaptive capacity of the placenta under RUPP procedure. Initial findings at GD14 showed significantly lower perfusion parameters in the RUPP compared to normal pregnancies. However, improvement by GD16 and GD18 suggests adaptive response that restore function over time. This result underscores the limited critical intervention window in the RUPP animal model and it provides insights into placental resilience and adaptation.
Fatimah AL DARWISH (Amsterdam, The Netherlands), Caren VAN KAMMEN, Lindy ALLES, Fieke TERSTAPPEN, Caren VAN KAMMEN, Raymond SCHIFFELERS, Titia LELY, Gustav STRIJKERS, Bram COOLEN
14:00 - 15:30
#47871 - PG392 MACHINE LEARNING-BASED CLASSIFICATION OF NEURO-BEHCET'S SYNDROME USING DIFFUSION TENSOR IMAGING EIGENVALUE METRICS.
PG392 MACHINE LEARNING-BASED CLASSIFICATION OF NEURO-BEHCET'S SYNDROME USING DIFFUSION TENSOR IMAGING EIGENVALUE METRICS.
Neuro-Behçet’s syndrome (NBS) is the neurological form of Behçet’s disease. Machine learning (ML) and artificial intelligence (AI) are increasingly used in medical imaging to enhance diagnosis and prediction, particularly in radiology. MRI provides both traditional and advanced biomarkers—such as lesion characteristics and diffusion-based metrics—that help quantify brain changes. These biomarkers support predictive modeling even in rare conditions like NBS. Combining multimodal imaging with ML offers potential for personalized prognosis.
Most research compares NBS patients to BS patients, with limited direct comparisons to healthy controls. A recent 2024 study [9] analyzed DTI data from 12 NBS patients and healthy controls, highlighting WM differences. This current study is notable for its relatively large NBS cohort from Türkiye, focusing on WM abnormalities using diffusion anisotropy indices and applying ML to identify key DTI metrics for accurately distinguishing pathological from healthy tissue.
Data were collected as a part of YOK 50006 "ML Based Differentiation of NBS with multi contrast MRI" project. project. This IRB approved study involved 14 NBS patients and 38 healthy controls, excluding one patient with a large frontal lesion. High-resolution T2-weighted MRI and diffusion tensor imaging (DTI) with 20 gradients were acquired. Three eigenvalue maps (λ₁, λ₂, λ₃) were generated.
White matter (WM) masks were semi-automatically segmented from T2-weighted images using MIPAV’s level set method, starting at the axial slice where lateral ventricles appeared to reduce non-WM inclusion. T2W and B0 images were skull-stripped, bias-corrected, and co-registered via affine transformations using ANTS, with WM masks transformed accordingly and binarized.
WM masks were semi-automatically segmented from T2-weighted brain MRIs using MIPAV’s level set method. Volumes of Interest (VOIs) were selected for both NBS and control groups, beginning segmentation at the axial slice where the lateral ventricles first appeared. This approach minimized non-WM inclusion, thereby reducing errors in diffusion tensor analysis.
Voxel-wise WM diffusion indices—including λ₁, λ₂, λ₃, ADC, RD, FA, and RA—and their means were calculated. Min-max normalization was applied to histogram-based features (30 bins) of these variables to ensure comparability. These features served as inputs for classification.
ML models were trained and tested using MATLAB’s toolboxes with leave-one-out cross-validation, evaluating features alone and in combination, with and without PCA (set to 5 components and 95% variance explained). ANOVA guided feature selection. Model performance was assessed via accuracy and ROC curves. The study found that λ₁ and λ₂ was the most sensitive markers. ADC’s importance is driven by λ₁ and λ₂, while RD is influenced by λ₃, which is less distinct. FA and RA showed no significance, indicating that raw diffusivity metrics better detect WM abnormalities in NBS.
Classification results demonstrated that λ₂, λ₃, and ADC achieved the highest accuracies, frequently exceeding 90%, with top performances reaching 98.08% for ADC (e.g., Fine Tree classifier). Quadratic SVM and Neural Networks also showed strong results, with accuracies up to 96.15%–100% after PCA application. PCA generally improved classification accuracy by 2–5%. In contrast, FA and RA consistently yielded lower accuracies, typically below 80%. These findings underscore λ₁, λ₂, λ₃, and ADC as the most sensitive and discriminative metrics for differentiating NBS patients from healthy controls. The 100% accuracy of the best model was shown in Figure 4. The analysis revealed that traditional metrics (FA, RA) lacked significance for NBS, whereas raw eigenvalue features particularly ADC were highly effective. ADC alone reached up to 100% accuracy with Linear SVM and neural networks. Combining ADC with other metrics, especially using PCA, further enhanced performance. Ensemble methods, SVMs, and neural networks consistently showed strong results, with Linear SVM notably efficient. Despite some model instabilities, ADC proved crucial for accurate WM integrity classification and clinical application. The analysis showed that traditional metrics like FA and RA were not significant for NBS, while raw eigenvalue features—especially ADC—were highly effective. ADC alone achieved up to 100% accuracy with models like Linear SVM and neural networks. Combining ADC with other metrics improved performance, especially with PCA. Ensemble methods, SVMs, and neural networks consistently performed well, with Linear SVM being notably efficient. Some models failed due to instability, but overall, ADC proved central for robust WM integrity classification and clinical potential.
Statistical analysis reveals group differences, but lacks predictive power. ML enables individual-level classification by modeling multiple features, especially in high-dimensional medical imaging data like DTI.
Yıldız TUZUN (Türkiye, Turkey), Hamit Alp COMERT, Irem CEVIKCAN, Merve BAYER, Adeola ADEREMI, Abdullah ARCAN, Ugur UYGUNOGLU, Alp DINÇER, Aksel SIVA, Alpay OZCAN
14:00 - 15:30
#47722 - PG393 Mood induction by reliving memories – impact on mood state and cerebral perfusion.
PG393 Mood induction by reliving memories – impact on mood state and cerebral perfusion.
Arterial Spin Labeling (ASL) perfusion MRI is a promising tool for detecting early biomarkers of neurodegenerative and psychiatric diseases [1]. However, significant physiological variability in cerebral blood flow (CBF) complicates its clinical use. Changes in mood may contribute to this variability and confound the interpretation of CBF measurements. This study evaluates the impact of mood on CBF using autobiographical recall with guided imagery during ASL scans.
ASL data in 48 healthy young adults (50% women, 18–29 years) were acquired during positive, negative, and neutral mood conditions, using a within-subject design (Figure 1). Mood was induced through guided imagery using autobiographical recall, trained before the MRI session. Perfusion was measured using a pseudo-continuous ASL with single-shot 3D-GRASE readout (labeling duration 1.5s, PLD 2s) on a single Siemens 3T Trio scanner at the UGent core facility GIfMI. ASL was processed using ExploreASL [2] and CBF was quantified for total gray matter (GM), anterior cingulate cortex (ACC), posterior cingulate cortex (PCC), hippocampus, anterior and posterior parahippocampal gyrus, amygdala, medial (pre)frontal cortex (mPFC) and putamen. Using linear mixed-effects models (LMM), the validity of the mood induction procedure, the GM CBF and spatial COV, and normalized regional CBF were tested for all regions. Multiple comparisons were corrected using the false discovery rate correction (FDR), and the significance level was set at alpha = 0.05. Mood induction effectively altered self-reported happiness and sadness, with significant task-valence interactions for happiness (F(2,811)=166.38, p<0.001) and sadness (F(2,810.01)=217.67, p<0.001). A significantly higher total GM CBF for positive MIPs (mean(SE) = 0.8 (+-0.3) ml/100g/min) compared to negative MIPs (t(811)=2.62,p=0.025), but not neutral MIPs (t(811)=2.25,p=0.063) was detected. Spatial COV results showed no significant effects (F′s<2.31,p′s>0.099). For regional perfusion, no significant main effect of mood state was detected (F’s < 1.04, p’s > 0.247). Task effects (during mood induction versus during resting-state) were observed for PCC (F(1,811)=4.77,p=0.029) and mPFC (F(1,811)=5.18,p=0.023), though these became non-significant after correcting for multiple comparisons. Mood changes were successfully induced during the MRI session. Positive mood induction significantly increased total GM perfusion compared to negative mood, while this effect was not detected for regional perfusion. However, compared to other perfusion modifiers such as age, exercise and sleep [3], these effects are relatively small. In conclusion, a positive mood appears to significantly influence total GM perfusion. Albeit small effect size, mood state may influence cerebral blood flow and should be considered in future perfusion studies.
Patricia CLEMENT (Ghent, Belgium), Naomi VANLESSEN, Stefanie DE SMET, Laura VANSTEENKISTE, Soetkin BEUN, Pieter VANDEMAELE, Stephanie BOGAERT, Henk-Jan MUTSAERTS, Gilles POURTOIS, Eric ACHTEN
14:00 - 15:30
#47730 - PG394 Influence of pulmonary transit time methods on discrimination between amyloidosis and normal heart.
PG394 Influence of pulmonary transit time methods on discrimination between amyloidosis and normal heart.
Pulmonary transit time (pTT) determined by cardiac magnetic resonance (CMR) has been shown to be impaired by various conditions such as heart failure [1], congenital heart disease [2] or surgical cardiac procedures [3]. Recently, pTT was also suggested as imaging predictor for cardiac involvement and prognosis in light-chain amyloidosis [4]. Frequently, pTT is determined from first-pass perfusion CMR as the time interval between the maxima of time-intensity curves in the right ventricle (RV) and left ventricle (LV). However, it has been suggested by Nelsson et al. [5] that the use of the center of gravity of the respective time-intensity curves might be more robust. It was the purpose of our study to compare the maximum-method and the center-of-gravity method with regard to discrimination between normal patients and patients with proven amyloidosis.
The study was approved by our local institutional review board. Patients (age > 18a, no contraindications for MRI) with proven amyloidosis and individuals without amyloidosis and no signs of cardiac impairment were included into this study. Subjects were investigated on a 1.5T MR scanner (MAGNETOM AvantoFit, Siemens, Germany). For pTT assessment, a first-pass perfusion sequence was applied, using a cardiac-gated single-shot 2D saturation recovery gradient echo sequence (true fast imaging with steady-state free precession - trueFISP); typical imaging parameters were as follows: repetition time/echo time: 2.2 ms/0.97 ms, saturation recovery time: 90 ms, flip angle: 50°, acquisition matrix: 128 × 82, field of view: 400 × 300 mm, bandwidth: 1370 Hz/pixel, slice thickness: 8 mm, parallel imaging mode: GRAPPA, and acceleration factor: 2. Image acquisition was started simultaneously with contrast media injection; a total of 60 images in one 4-chamber long-axis view as well as two short-axis views were acquired.
Time-intensity curves were created with syngo.via software (VB80f, Siemens Healthineers) and circular regions of interest (ROI) were placed within the basal RV and LV blood pool on the first-pass perfusion 4-chamber long-axis view. Care was taken to avoid partial volume effects at the papillary muscles or myocardial wall. Average signal intensity within the ROI was plotted as a function of time, resulting in an indicator-dilution curve. Omitting recirculation, a gamma variate fit was applied to the data using the R Project for Statistical Computing 4.4.3 software (R Foundation for Statistical Computing). From the obtained fit parameters time to peak values (TTP) and time to center of gravity (CoG) values could easily be computed. pTT was then defined as the difference between the respective LV and RV values. 114 subjects were enrolled in this study, including 19 subjects, which were chosen as controls, without cardiac impairment and 89 patients with proven transthyretin amyloidosis (ATTR). Table 1 shows a summary of the obtained pTT values for heart-healthy subjects and ATTR amyloidosis patients for the different pTT methods. Wilcoxon rank sum test showed a significant difference between controls and patients with amyloidosis for pTT values of both methods (fig 1). ROC analysis resulted in similar pTT thresholds for both methods (9.55s for pTT(TTP) and 9.57s for pTT(CoG)). The area under the curve (AUC) for pTT(TTP) was 0.907 (95% CI: 0.849-0.965) with a specificity of 100% and a sensitivity of 71.9%. For pTT(CoG) the AUC was 0.668 (95% CI: 0.552 – 0.783) with a specificity of 84.2% and a sensitivity of 57.3%. As shown in figure 2, the difference between both ROC curves was highly significant (p < 0.001). In this study pTT calculated by two different methods from first pass contrast enhanced CMR data were compared between controls and patients with proven ATTR amyloidosis. pTT(TTP) enabled a significantly better discrimination between the patient groups as compared to pTT(CoG). For pTT(CoG), variablity in contrast agent wash-out may lead to the observed decrease in discrimination between the patient groups. pTT(TTP) obtained as difference between LV and RV TTP values were found to enable a significantly better discrimination between heart-healthy subjects and ATTR amyloidosis patients.
Christian KREMSER (Innsbruck, Austria), Philip LUNGENSCHMID, Felix TROGER, Maria UNGERICHT, Agnes MAYR
14:00 - 15:30
#47740 - PG395 Structural characterization of cardiac Purkinje fibers using optimized inhomogeneous Magnetization Transfer (ihMT) MRI and Histology.
PG395 Structural characterization of cardiac Purkinje fibers using optimized inhomogeneous Magnetization Transfer (ihMT) MRI and Histology.
The His-Purkinje system is implicated in the initiation and maintenance of ventricular fibrillation, which can lead to sudden cardiac death [1]. This conduction network is essential for ventricular activation but remains poorly characterized and structurally variable across species [2].
Inhomogeneous Magnetization Transfer (ihMT) MRI [3–5], sensitive to myelinated structures [6], has recently shown promise for imaging the cardiac conduction system, particularly Purkinje fibers [7–9]. This study aims to optimize ihMT parameters for PF visualization, and complement it by histological validation.
Experimental setup:
A left ventricular sample (Fig.1-a), containing myocardium and free-running PF was obtained from a female sheep’s (51.1 kg, 9 y.o) heart and fixed in 4% formaldehyde containing 0.1% of gadoterate meglumine (0.5 mmol/mL; Dotarem, Guerbet, France). For MRI, the sample was placed in a syringe filled with Fluorinert (Sigma-Aldrich; Fig.1-a), heated to 33±1 °C using water bath and continuously monitored with a temperature probe (SA Instruments, NY).
MR acquisition & post-processing:
Experiments were conducted on a 9.4T/30cm (Bruker Biospin MRI, Ettlingen, Germany) with a cylindrical transmit coil (87-mm intern) coupled with a 4-channel phased-array reception coil. A 2D ihMT-prepared RARE sequence (Fig.1-b) was used with the following readout parameters: TE/TR/PF/#M0/#MTw/TA=20ms/3000ms/1.8/10/200/2h and res=0.25×0.25×1 mm3. IhMT ratios (ihMTR%) were calculated as:
ihMTR=100×(M_sing-M_dual)/M_0
where Msing=MT++MT- and Mdual=MT±+MT∓.
MT+ and MT- refer to the MT-weighted images acquired with positive frequency offset saturation and negative offset saturation, respectively and MT± and MT∓ refer to MT-weighted images acquired with dual frequency offset saturation. IhMTR maps were generated using the script: https://github.com/lsoustelle/ihmt_proc. A range of different MT saturation parameters (Table 1) were tested to find the optimized contrast defined as:
Contrast=ihMTR_PF - ihMTR_myocardium
where a custom-made python script was used to draw two ROIs manually in PF (Fig. 1c, Mask1, red) and in myocardium (Fig. 1c, Mask1 green) to calculate ihMTR mean±SD. Further analysis was done by separating PF into free-running (Fig. 1c, Mask2, red) part of the fiber and the insertion point (Fig. 1.c, Mask2, blue), the part that enters the myocardium.
Histological analysis:
After MR experiments, histological analysis was conducted to provide detailed insights into the tissue structure. After dehydration, sample was embedded in paraffin and sectioned at 6 μm. Tissue sections were stained with Picro-Sirius Red for structural identification: collagen fibers, myocytes, and adipocytes were red, yellow and white, respectively. The polarized light (PL) filter (analyzer-polarizer, Nikon) allowed for differentiation between collagen type I (in red and yellow) and collagen type III (in green) [10]. Figure 2 illustrates ihMTR values in the PF (red) and myocardium (green) across different experiments. In all acquisitions, ihMTR was consistently higher in PF compared to myocardium. The protocols with the highest contrasts (namely, Pr1=2±1.2%, Pr2=2.1±1.3%, and Pr3=2±1.2%) are highlighted in the figure (dashed lines), and their corresponding MT saturation parameters are listed in the table.
ihMTR values across the three protocols consistently showed higher signal in the free-running region of the PF vs. the insertion point. In Pr1, the values were 9.4 ± 1.6% vs. 6.6 ± 1.8%; in Pr2, 11.0 ± 1.9% vs. 8.1 ± 1.2%; and in Pr3, 9.4 ± 1.8% vs. 6.9 ± 1.6% for free-running vs. insertion point. For comparison, myocardium ihMTR values were 7.2 ± 0.7% in Pr1, 8.6 ± 0.8% in Pr2, and 7.2 ± 0.7% in Pr3.
Fig. 3a shows histological views of the same sample under standard and polarized light (PL), along with the ihMTR map from Pr1 (Fig. 3b). The fiber contains Purkinje cells, collagen (red), and adipocytes (white), with adipocytes mainly at the insertion point and collagen more abundant in the free-running region. Under PL, Col I (red/yellow) is more prevalent than Col III (green); Col III is slightly more expressed at the insertion point. Testing different ihMT protocols on one sample identified those with highest fiber signal and myocardium contrast. IhMTR was higher in the free-running PF region, which showed more collagen and Purkinje cells. The insertion point had lower ihMTR, likely due to adipocytes. IhMT effectively detects microstructural differences in the Purkinje network linked to tissue composition. The Purkinje network is a variable and understudied structure. IhMT offers contrast sensitive to its heterogeneous makeup, influenced by collagen and adipocytes. This study establishes a framework for fiber imaging and validates it histologically. Future work will apply this approach to additional ex-vivo samples to better characterize ihMT contrast.
Arash FORODIGHASEMABADI (Bordeaux), Evgenios N. KORNAROPOULOS, Marion CONSTANTIN, Lucas SOUSTELLE, Fanny VAILLANT, Jude LEURY, Richard WALTON, Olivier BERNUS, Bruno QUESSON, Olivier M. GIRARD, Guillaume DUHAMEL, Julie MAGAT
14:00 - 15:30
#47561 - PG396 MRI-Based Radiomics for Survival Prediction and Risk Assessment in Glioma Patients.
PG396 MRI-Based Radiomics for Survival Prediction and Risk Assessment in Glioma Patients.
Glioma is the most common and aggressive primary cancer of the central nervous system[1][2]. Despite recent advances in clinical practice, the detection of prognostic biomarkers still requires invasive, complex, and costly methods[3][4]. Therefore, the need for a simple, reliable, and easy-to-use predictive model arises.
MRI is a noninvasive modality known for its superior soft tissue contrast, making it particularly useful for diagnosing brain pathologies, including glioma. Radiomics is a field within medical imaging that specializes in the analysis and extraction of quantitative parameters from medical images. Given these insights, radiomics features extracted from MR images can unveil hidden patterns that cannot be identified on other imaging modalities, serving as essential building blocks for the development of prognostic prediction models and treatment decision tools in cancers.
The goal of this study is to construct and validate radiomics prediction models based on preoperative gadolinium-enhanced T1- and T2-weighted images, and to evaluate their performance.
The UPENN-GBM (611 patients) and TCGA-GBM (135 patients) cohorts from TCIA (The Cancer Imaging Archive) were used in this study[5][6]. These datasets include magnetic resonance imaging (MRI) scans of de novo glioblastoma patients with gadolinium-enhanced T1-weighted, T1-weighted, T2-weighted, and FLAIR images. Automatic and manually corrected segmentations of tumor sub-regions—tumor core, enhancing tumor, and invasion—were available for all patients. UPENN-GBM was used for training, while TCGA-GBM served as an independent test set.
Radiomics features were extracted from three tumor sub-regions (tumor core, enhancing tumor, and invasion), using gadolinium-enhanced T1 and T2-weighted MRI, processed with PyRadiomics v3.1.0[7].
For voxel-wise radiomics, K-means clustering was performed on the features with k = 3, aiming to segment the tumor into distinct imaging habitats. Habitat imaging refers to the identification of spatially heterogeneous subregions within a tumor that may reflect underlying differences in biological characteristics such as cellularity, vascularity, or hypoxia. These radiologically defined habitats provide a noninvasive means to capture intratumoral heterogeneity and are increasingly used to guide personalized treatment strategies[8].
The number of clusters was based on the three tumor sub-regions, and the area of each cluster was calculated in cm³. Pearson correlation analysis was applied to feature pairs, and one from each pair with r > 0.9 was removed to reduce multicollinearity. The remaining features, along with patient age, were used in a Cox proportional hazards regression model.
All combinations of two and three predictors were tested. Four-fold cross-validation was used for robustness, and models were evaluated on the test set. Performance was assessed using P-values and the concordance index (C-index). Model simplicity was prioritized to avoid overfitting. As representative example, the clustering result of the same radiomics feature for two different patients is shown in Figure 1.
Thousands of models were built, trained, and tested based on the remaining features after applying Pearson correlation analysis. The C-index for the best models ranged from 0.682 to 0.689 (95% CI: 0.534–0.751; P < .0001) in conventional radiomics and from 0.665 to 0.667 (95% CI: 0.530–0.753; P < .0001) in spatial radiomics, with all models consistently based on three predictors. In conventional radiomics, the most predictive model was derived from features extracted from the invasion sub-region using T2-weighted images (Figure 2). Meanwhile, in spatial radiomics, the best predictive model utilized features from the whole tumor, extracted from gadolinium-enhanced T1-weighted images (Figure 3). Both radiomics approaches offered comparable results, with the classical method having a slight edge over the spatial radiomics approach, and the same can be said for the two weighting mechanisms. However, there were differences in model performance when features were extracted from different tumor sub-regions, primarily due to tumor heterogeneity. Although these results could be further improved by including more predictors in the model, our goal was to keep the model as simple as possible, which will facilitate the reproducibility and generalization of the results.
The next steps will be to improve model performance by implementing the elbow method to choose the optimal K-value for clustering, including more clinical information in the prediction model, and obtaining a second testing set from a local cancer treatment center in Lyon, France. The implementation of radiomics in clinical practice can be helpful for survival prediction and risk stratification. Both classical and spatial radiomics can offer a simple, non-invasive and easy to use tool to help clinician in their decision making
Walid DANDACHLY (Lyon), Benjamin LEPORQ, Frank PILLEUL, Vincent GREGOIRE, Olivier BEUF
14:00 - 15:30
#47772 - PG397 Sex-Dependent Effects of Obesity on Glioblastoma Features in a Murine Comorbidity Model: Insights from Multiparametric MRI.
PG397 Sex-Dependent Effects of Obesity on Glioblastoma Features in a Murine Comorbidity Model: Insights from Multiparametric MRI.
Obesity is a complex, chronic condition influenced by sex and associated with alterations in brain microstructure, which can be non-invasively assessed using magnetic resonance imaging (MRI) [1]. While obesity has been linked to an increased risk of several cancer types [2], its specific role in the progression of brain tumors—particularly glioblastoma (GBM), the most aggressive primary brain tumor in adults—remains unclear. The aim of this study is to investigate MRI biomarkers that reflect the impact of high-fat diet (HFD) exposure at 10 and 20 weeks on GBM characteristics in a preclinical mouse model, while also assessing potential sex-dependent differences.
Adult C57BL/6 mice were randomly assigned to one of four groups based on diet type and duration: high-fat diet (HFD, 60% fat) or standard diet (SD), for either 10 weeks or 20 weeks (around 9 mice per sex, diet and diet duration group). Following the respective dietary period, GBM was induced in all animals via stereotactic injection of 10⁵ GL261 cells into the brain parenchyma. GBM development was followed-up by T2W MRI on a 7T system. Once tumor volume reached approximately 70 mm³, a multiparametric MRI protocol was conducted, including T2 and T2* mapping, magnetization transfer ratio (MTR), and diffusion tensor imaging (DTI). Parametric maps were generated with Resomapper, a custom-made Python toolkit, and four regions of interest: tumor core (TC), tumor periphery (TP), peritumoral zone (PZ), whole tumor (WT) and contralateral apparently healthy brain (HB) were manually segmented and quantified using ImageJ. Linear mixed effects models were used with R to test for the effect of area, diet, diet duration and sex and the interactions between them. Post-hoc tests were performed in case any of these were found, using FDR correction for multiple comparisons. In all parametric maps, expected differences between regions of interest were observed. The HB area showed higher MTR and fractional anisotropy (FA) than the tumoral areas, and oppositely, higher values of T2, mean, radial and axial diffusivity (MD, RD, AD) were found in the tumor than in HB. T2* was more homogeneous, but slightly higher in the HB (Fig. 1).
Regarding DTI parameters, in MD a significant effect of the interaction Area:Diet:Duration (p<0.05) was observed. No significant differences between diets or timepoints were directly found, but differences between areas vary depending on the diet group. Particularly, in the 10 weeks HFD group, regardless of sex, TP showed a significant difference with the TC that does not appear in other groups that even showed the opposite tendency (Fig. 2). The results in RD are similar to these.
In AD only a Sex:Area effect was observed (p<0.05). Males tended to have higher AD values than females across all brain areas, specifically in the HB area, but differences were not statistically significant. Similarly, in FA some effects including sex were found but do not translate into significant group differences.
T2 results showed many significant effects, including a strong effect of the Sex:Diet:Duration interaction (p<0.001) and also of Area:Diet:Weeks (p<0.05). At 10 weeks, there is a significant difference between diets in the WT area, as well as in the TP, but only in females. Specifically, tumor of HFD females had higher T2 values than the corresponding controls. At 20 weeks, this difference disappeared in the females but appeared in males. Also, a difference between diets in the HB area was observed only in males, being the opposite at 10 weeks than at 20 weeks: T2 values were higher in HB of the SD mice at the early stage and higher in HFD mice at 20 weeks of diet (Fig. 3).
In T2* and MTR some significant effects including diet type and duration were found, but do not translate into significant differences between groups. First of all, differences in all parameters between the HB and tumoral areas are the expected, indicating presence of edema and haemorrhage in all tumor areas, and also slight edema in the PZ [3]. The most interesting insights come from MD and T2. MD suggests that in the 10-weeks HFD mice the edema is more concentrated in the TP respect the TC, which does not happen in other groups. Moreover, the T2 results indicate that the evolution with obesity stage of the water content of the tumor happens differently between sexes, and in the case of males also of the HB area, which might be affected by the tumor differently. Further work is currently being performed to validate these results with immunochemistry markers of neuroinflammation and proliferation, as well as 1H HRMAS spectroscopy. This work characterizes a murine model of GBM and obesity comorbidity, revealing that tumor characteristics and its effects on the rest of the brain are influenced by sex, diet type, and diet duration, despite numerous confounding factors. The immunochemistry and metabolomic spectroscopy assays will help understand the underlying mechanisms of these MRI results.
Raquel GONZÁLEZ-ALDAY, Nuria ARIAS-RAMOS, Blanca LIZARBE, Pilar LÓPEZ-LARRUBIA (Madrid, Spain)
14:00 - 15:30
#45869 - PG398 Investigating the dosimetric impact of gynecological brachytherapy applicator reconstruction on T2-weighted and T1-weighted MRI images versus CT images, and quantifying potential benefits of reconstructing on T1- versus T2- weighted images.
PG398 Investigating the dosimetric impact of gynecological brachytherapy applicator reconstruction on T2-weighted and T1-weighted MRI images versus CT images, and quantifying potential benefits of reconstructing on T1- versus T2- weighted images.
Brachytherapy (BT) for gynecological cancer treatment planning traditionally employs CT images for applicator and catheter reconstruction, and MRI imaging for organ and target contouring. Many centers now employ MRI-only BT planning. Reconstructing on MRI images poses challenges as applicators and catheters are more difficult to visualize on MRI than CT, and inherent MRI distortions can lead to dosimetric effects of 2-7% per mm of displacement. This research retrospectively assessed the dosimetric differences for gynecological brachytherapy treatment plans using applicators reconstructed on CT versus MRI images. Images from two MRI pulse sequences were evaluated: a T2-weighted-2D-PROPELLER and a T1-weighted-3D-LAVA-FLEX sequence.
The study included 12 cervical cancer patients undergoing three fractions each of BT with a Venezia (Elekta) applicator, either with or without catheters. Images from each implant were acquired on a 1.5 Tesla Avanto fit GE scanner and a Philips Big Bore Radiation Therapy CT scanner. The MRI scans consisted of a T2-2D-PROPELLER sequence and a T1-3D-LAVA-FLEX sequence. The MRI images were oriented in the plane of the tandem to minimize distortions.; thus, MIM (Medical Image Merge, version 7.2.8) was employed for reorientation. The Oncentra Treatment Planning System (Elekta, version 4.6.2) was used to reconstruct the applicators and catheters on each CT or MRI image. A 3D dose grid was calculated in Oncentra for each MRI or CT reconstruction using the dwell times and dwell positions from the clinical treatment plan. A gamma index analysis was employed to compare the dose maps between the reconstructions done on CT and MRI images, using a passing rate of 90% with a 3%/2mm dose difference/distance to agreement, and 10% maximum dose thresholding. Average gamma results were obtained for the T2- and T1-weighted images. A Wilcoxon signed ranked test was completed (significance level of 0.05, two-tailed) to assess if measured distortions using the T2-2D-PROPELLER versus the T1-3D-LAVA FLEX reconstruction are significantly different. Moreover, we investigated if the number of catheters affected gamma pass rates. A Wilcoxon rank-sum test was used to assess if one sequence outperformed the other for each number of catheters. In some cases, the MRI sequences being investigated were not completed due to logistical issues. These cases were removed from the study. The T2- and T1-weighted images from two different studies were not included due to image corruption issues. In total, 27 T2-weighted images and 27 T1-weighted images were included. Figure 1 displays the reconstruction in example CT, T2-weighted, and T1-weighted images for a patient. For the Wilcoxon signed rank test comparing gamma results obtained from T2-weighted images to T1-weighted images, 26 total samples were included since the unusable images mentioned above were from different cases. Of these cases, 19 included catheters and the rest were applicator only. The average gamma result over all T2- and T1-weighted image reconstructions were 96.1 ± 2.8% and 96.6 ± 3.1%, respectively. Minimum gamma results were 90.2% and 90.5% for T2- and T1-weighted images, respectively. Figure 2 displays the skew of gamma pass rates for the T2- and T1-weighted images. Figure 3 reveals the gamma analysis results for T2-2D-PROPELLER and T1-3D-LAVA-Flex reconstructions versus the number of catheters. The Wilcoxon tests indicated there was no significant difference between T2- and T1-weighted reconstructions overall, or for 0 to 5 catheters. However, T1-3D-LAVA-FLEX significantly outperformed T2-2D-PROPELLER for cases with 6 catheters. Figure 1 reveals that the T2-PROPELLER and T1-3D-LAVA-FLEX MRI images are of sufficient quality for reconstruction and contour delineation. All images passed the gamma index criterion of 90%. The results illustrate that there isn’t a significant difference between dose maps obtained from applicator reconstructions on T2-2D-PROPELLER and T1-3D-LAVA-FLEX MRI images compared to CT images. However, as seen in Figure 2, reconstructions done on T1-3D-LAVA-FLEX are skewed towards higher gamma rates compared to those done on T2-2D-PROPELLER. Notably, Figure 3 reveals that the mean T2-2D-PROPELLER minus T1-3D-LAVA-Flex gamma differential is within the standard deviation of the gamma pass rates (~ 3%) when 0 to 4 catheters are reconstructed. As catheter number increases to 5 or 6, T1-3D-LAVA-Flex reconstructions perform superiorly to T2-2D-PROPELLER reconstructions. T1-weighted images significantly outperform T2-weighted images for 6 catheters. These results suggest that T1-weighted images may be beneficial for image reconstruction, particularly if using more catheters. T2-2D-PROPELLER and T1-3D-LAVA-FLEX MRI images are safe and effective for MRI-only gyne bracytherapy. T1-weighted images may offer benefits for reconstructions of 6 catheters or more.
Clara J FALLONE (Calgary, Canada), Matthew J FRICK
14:00 - 15:30
#47297 - PG399 Intraoperative arterial spin labeling in pediatric posterior fossa brain tumors – methodological implications.
PG399 Intraoperative arterial spin labeling in pediatric posterior fossa brain tumors – methodological implications.
Central nervous system tumors are the most common solid tumors in children, with about half arising in the posterior fossa [1,2]. Following surgery, approximately 25% of children with posterior fossa tumors develop cerebellar mutism syndrome (CMS) - a condition marked by delayed-onset speech, motor, and emotional disturbances typically 24-48 hours postoperatively [3,4]. Early identification of anatomical or functional changes, such as cerebral blood flow (CBF), is critical for understanding the pathophysiology of CMS. Intraoperative MRI provides a unique opportunity for monitoring of CBF during surgery.
Arterial spin labeling (ASL) is a non-invasive MRI technique used to quantify CBF and is implemented during intraoperative MRI procedures at our center. However, CBF measurements can be influenced by various factors, including anesthesia, patient positioning, and MR acquisition parameters. To examine the methodological implications for interpreting intraoperative CBF values, we compared ASL scans acquired during surgery to those obtained the day prior in children with posterior fossa tumors.
Study design
Six patients (2m/4f; age: 6.7±4.9 years) who underwent surgical resection for posterior fossa tumors were included. Written informed consent was obtained.
Images were acquired using a 3-Tesla MRI scanner (Ingenia ElitionX, Philips Healthcare). Preoperative scans (timepoint 1) were obtained prior to surgery (2.8±2.7 days), using a 32-channel head receive coil with the patient positioned in supine position. Intraoperative scans (timepoint 2) were conducted in the operating room (prior to craniotomy), with patients positioned in surgical prone position, secured in a head clamp, and scanned using two single-loop RF coils to accommodate the intraoperative setting (Figure 1).
ASL acquisition
Pseudo-continuous ASL scan protocols were consistent across both timepoints, utilizing a 3D GRASE sequence with a labeling duration of 1800 ms and a post-labeling delay of 1800 ms. Imaging parameters were as follows: 8 dynamic scans; 4 background suppression pulses; matrix=64x64x23; FOV=240x240x161 mm3; acquired voxel size=3.75x3.75x7.0 mm3; TE=13ms; flip angle=90°. Repetition time (TR) differed between the two timepoints, which was 4280 ms at timepoint 1 and 4600 ms at timepoint 2.
Image processing
Quantitative CBF maps were generated using established recommendations [5]. The Bayesian Inference for Arterial Spin Labeling (BASIL) toolbox [6] was used to compute CBF values (mL/100 g/min). Inversion efficiency was set to 0.69 and T1 of blood was adjusted using patient-specific hematocrit values [7]. Adaptive spatial regularization and motion correction were applied, and voxel-wise calibration using M0 images was performed.
To extract CBF values in cerebral gray matter (GM), tissue segmentation was performed on T1-weighted images using the FAST toolbox in FSL. GM maps were subsequently registered to native ASL space. Mean CBF values for cerebral GM were computed, and paired t-tests compared the mean cerebral GM CBF values between timepoint 1 and timepoint 2. CBF maps for two patients are presented in Figure 2, illustrating systematic differences in perfusion. Quantitative analysis revealed that mean CBF values in the cerebral GM at timepoint 1 (50.16±26.11) were significantly higher than those at timepoint 2 (14.40±5.76) (p=0.01; Figure 3). Notably, this pattern of decreased CBF was consistent across patients. In children with posterior fossa tumors, intraoperative ASL shows significantly lower CBF values compared to measurements obtained prior to surgery. This difference may arise from factors such as patient positioning (supine vs. prone). Studies in healthy adults have shown no significant CBF differences across these positions, except for a trend toward slightly lower CBF in prone position [8]. Similar comparisons have not been conducted in children.
Intraoperative ASL imaging is limited by image acquisition constraints, including alignment along the anterior commissure–posterior commissure line, which may hinder the labeling slab from being perpendicular to the arteries, particularly when patients are in prone position. Additionally, due to time constraints, angiography is not feasible, further limiting accurate positioning of the labeling slab. These limitations reduce inversion efficiency, potentially leading to underestimated CBF values.
These constraints complicate longitudinal CBF comparisons. Individual measurement of inversion efficiency, such as through phase-contrast velocity MRI [9], may help mitigate these effects. Alternatively, relative CBF maps can be used, although this will not preserve global CBF changes. In children with posterior fossa tumors, intraoperative CBF values are significantly lower than preoperative values, potentially making comparison of absolute CBF measurements less reliable. Methodological factors, especially inversion efficiency, may contribute to the observed CBF differences.
Iris OBDEIJN (Utrecht, The Netherlands), Rick BRANDSMA, Thomas LINDNER, Pien JELLEMA, Eelco HOVING, Marita PARTANEN
14:00 - 15:30
#45916 - PG400 Voxel-Wise Assessment of Tumor Hypoxia in a Murine Pancreatic Cancer Model Using TOLD-MRI and Histological Correlation.
PG400 Voxel-Wise Assessment of Tumor Hypoxia in a Murine Pancreatic Cancer Model Using TOLD-MRI and Histological Correlation.
Pancreatic cancer is one of the most aggressive cancers and tumors are characterized by hypoxia [1]. Hypoxia induces numerous adaptive cellular responses that contribute to tumour resistance to radiotherapy and certain drugs, and is a marker of aggressiveness [2]. To noninvasively map tumor hypoxia, several teams have explored MRI-based approaches, particularly Tissue Oxygen Level-Dependent (TOLD) contrast that uses oxygen as a contrast agent [3,4]. In this work, we attemp and validate by correlate on voxel wise with IHC marker CAIX. The aim of this study was to assess tumour hypoxia using TOLD contrast in mice subcutaneously grafted with pancreatic tumours, and to validate the method by voxel-wise correlation with reference histological imaging.
Two pancreatic cancer cell lines, AsPC1 and SW1990, were subcutaneously injected into the flanks of five mice per group. Once tumors reached 500 mm³, mice were anesthetized for MRI acquisition using a 9.4 T preclinical scanner. The center of each tumor was marked on the skin and overlaid with a water-filled tube to define the imaging plane (Fig. 1). The MRI protocol included a high-resolution T2w sequence, DW-MRI, and an inversion recovery fast spin echo multislice (IR-FSEMS) sequence. The IR-FSEMS sequence was acquired twice during air and twice during oxygen breathing for TOLD measurements. For TOLD analysis, R1 maps were generated by fitting IR-FSEMS data acquired under air (R1_air) and oxygen (R1_O2) conditions, and %∆R1 maps were computed as %∆R1=(mean(R1_O2) – mean(R1_air))/mean(R1_air). ADC maps were extracted from DW-MR images.
Following MR scans, mice were euthanized, and tumors were excised and sectioned parallel to the imaging plane (Fig. 1). From each tumor, six 2 µm-thick histological sections were obtained at 150 µm intervals to capture the full histological information corresponding to the MRI slice thickness. Slides were stained with CAIX IHC, a marker of hypoxia, and a threshold-based segmentation approach was applied using QuPath to identify hypoxic pixels based on positive staining. The resulting CAIX-classified images were interpolated to the MR resolution, generating hypoxic fraction maps and then registered to the MR images using manual affine and automatic non-rigid registrations. Hypoxic pixels were identified on %∆R1 and CAIX fraction maps using the following thresholds: %∆R1 ≤ 0 and CAIX fraction >0.15.; all other pixels were classified as normoxic. High ADC values were used to identify necrotic regions, which were excluded from the hypoxic pixel classification. Hypoxic segmentation maps derived from TOLD imaging were compared on a pixel-wise basis with those obtained from CAIX IHC. Evaluation maps were generated to assess the accuracy of TOLD-based classifications, distinguishing correctly identified normoxic and hypoxic pixels from misclassified ones. Figure 2 presents TOLD- (%∆R1) and CAIX- based segmentation maps alongside the corresponding evaluation maps for two tumors from each cell line (AsPC1 and SW1990).
Figure 3a presents the percentage of pixels from the evaluation maps, indicating the proportion of correctly and incorrectly classified hypoxic and normoxic pixels based on TOLD imaging, across both cell lines and all tumours. Figure 3b displays the number of pixels identified as hypoxic or normoxic by the TOLD-based classification compared to the reference classification derived from CAIX-stained histological images. In both MRI and histological assessments, the SW1990 cell line produced more homogeneous tumors, with a greater proportion of normoxic pixels, compared to AsPC1 tumors, which exhibited a higher prevalence of hypoxic areas. AsPc1 tumors displayed increased heterogeneity, with a more balanced distribution of hypoxic and normoxic pixels. This balance in AsPC1 tumors contributed to a higher classification error for TOLD imaging (51%) relative to SW1990 (29%), resulting in an overall error rate of 36%. Furthermore, the distribution of pixel counts across classification groups was consistent between TOLD and CAIX evaluations. This study successfully established a workflow for validating an MRI-based method against histological reference images on a voxel-by-voxel basis, despite a substantial difference in resolution (0.46x0.46x2 µm3 vs 250x250x1000 µm3). TOLD contrast proved effective in assessing hypoxia in subcutaneously grafted pancreatic tumors in mice, showing a strong correlation with histological findings. This approach enables the evaluation of therapeutic efficacy in pancreatic cancer by measuring hypoxia, providing a valuable tool for future drug efficacy studies.
Marion TARDIEU (Montpellier), Maïda CARDOSO, Tristan MANGEAT, Christophe GOZE-BAC, Bruno ROBERT, Véronique GARAMBOIS, Stéphanie NOUGARET, Christel LARBOURET
14:00 - 15:30
#47943 - PG401 Voxel-wise confidence range of DCE-MRI in breast cancer: A Monte Carlo pilot investigation of extended Tofts models.
PG401 Voxel-wise confidence range of DCE-MRI in breast cancer: A Monte Carlo pilot investigation of extended Tofts models.
Pharmacokinetic (PK) models, a quantitative method of DCE-MRI to offer quantitative physiological markers of plasma volume fraction (vp), extravascular extracellular space fraction (ve), and permeability–surface area product (PS), have been employed to facilitate tumour stratification and treatment planning [1–3]. Extended Tofts Models (ETM), a category of PK models, is consisted of Fast Exchange Limit (FXL) and No Exchange Limit (NXL) approaches with proven utility in specific clinical scenarios, however the suitability for breast cancer imaging is yet to be established. We therefore set out to investigate the suitability of FXL and NXL approaches for breast cancer imaging using Monte Carlo simulation, and to explore the determinants for voxel wise confidence range.
We hence acquired DCE-MRI from a patient with invasive ductal carcinoma and conducted FXL and NXL analysis, and numerical simulation to derive voxel wise coefficient of variation (CV), with study design shown in Figure 1. The study was approved by the London Research Ethics Committee (Identifier: 17/LO/1777) and registered as a clinical trial [NCT03501394].
Imaging Experiment: DCE-MRI data were acquired on a 3T MRI scanner (Achieva TX, Philips Healthcare, Best, Netherlands), using a 3D T1-weighted spoiled gradient echo (SPGR) sequence, with a repetition time (TR) of 3.8 ms, echo time (TE) of 2.3 ms, flip angle of 12˚, voxel size of 1.0 × 1.0 × 1.5 mm³, and 29 dynamics. Image analysis was conducted using MRIcron (University of South Carolina, USA), with rigid-body motion correction applied.
Maps of vp, PS, ve were computed separately for FXL and NXL analysis using SEPAL algorithm [4], with model T1 of 1.3 s [5], r₁ relaxivity of 5.9 s⁻¹·mM⁻¹, r₂ relaxivity of 17.5 s⁻¹·mM⁻¹ [6] and a population-averaged arterial input function (AIF) [7].
Numerical Simulation: The perfect signal for a voxel was generated using corresponding experimentally derived vp, PS, ve from FXL analysis. The baseline signal was computed as the average of the corresponding first 4 time points, and subsequently the time course of each voxel was normalised as the percentage difference of the baseline signal. The experimentally derived SNR of the same voxel from FXL analysis was employed to generate Gaussian-distributed noise and added to the perfect signal to simulate the realistic signal. The realistic signal was analysed using the FXL approach to yield simulation derived vp, PS, ve. The noise generation and fitting were repeated 100 times to create 100 sets of outputs and subsequently the mean, standard deviation and CV were computed. Maps of mean, standard deviation and CV for vp, PS, ve were obtained by iterating the simulation through all the voxels within the tumour. Subsequently, the maps were computed for NXL using experimentally derived vp, PS, ve and SNR and NXL analysis for fitting.
Statistical Analysis: All statistical tests were conducted using SPSS (Release 29.0, SPSS Inc., Cincinnati, OH, USA). Wilcoxon signed-rank paired tests were performed to evaluate the differences in CV between FXL and NXL analysis. Spearman’s correlations tests were performed for CV against SNR and mean values. There is no significant difference in vp, PS, ve between FXL and NXL analysis, and all the statistical findings can be found in Table 1.There was a significant difference in CV of vp (p < 0.001, Figure 3A) between FXL (0.0107 ± 0.1213) and NXL (0.0111 ± 0.1227). There was a significant difference in CV of PS (p < 0.001, Figure 3A) between FXL (0.0032 ± 0.1159) and NXL (0.0146 ± 0.2253). There was a significant difference in CV of ve (p < 0.001,Figure 3A) between FXL (0.0064 ± 0.0485) and NXL (0.0181 ± 0.1144). There was a significant negative correlation between CV of vp (p < 0.001), PS (p < 0.001), ve (p < 0.001) from FXL against SNR (Figure 3B). There was also a significant negative correlation between CV of vp (p < 0.001), PS (p < 0.001), ve (p < 0.001) from FXL against corresponding mean value (Table 1). Although the mean PK model outputs showed no significant difference between FXL and NXL models with reasonable number of instances, the CV of FXL is significantly lower than NXL on all the output parameters, indicating FXL as the preferred model for measurement error consideration. Strong negative correlations between CV and both SNR and central parameter values indicate that signal quality and parameter magnitude jointly influence uncertainty. These findings highlight the importance of model choice and SNR-aware interpretation in voxel-level DCE-MRI analysis. Voxel wise Monte Carlo simulations enable spatially resolved confidence range quantification of DCE-MRI in breast cancer. The FXL model showed lower variability compared to NXL model.
Rachaita PODDER (Newcastle Upon Tyne, United Kingdom), Sai Man CHEUNG, Kangwa NKONDE, Andrew BLAMIRE, Jiabao HE
14:00 - 15:30
#47909 - PG402 Hypoxia-Targeted BOLD MRI For Differentiating True Progression From Pseudoprogression/ Radionecrosis In Glioblastoma: A Pilot Study.
PG402 Hypoxia-Targeted BOLD MRI For Differentiating True Progression From Pseudoprogression/ Radionecrosis In Glioblastoma: A Pilot Study.
Distinguishing true glioblastoma progression/recurrence from treatment-related pseudoprogression/radionecrosis is a clinical challenge.[1] Conventional MRI routinely cannot differentiate between these entities, resulting in a risk of overtreatment or delay of necessary interventions.[2] Hypoxia targeted blood oxygen level-dependent (BOLD) MRI is a novel imaging technique that may allow for enhanced characterization of tumor tissue based on underlying vascular features and oxygenation metabolism. In this study we prospectively aim to test the hypothesis that the information provided by hypoxia-targeted BOLD MRI may help differentiate between tumor progression/recurrence and treatment effect changes.
Patients with radiographically suspected glioblastoma progression/recurrence were included to undergo a hypoxia targeted BOLD MRI. A computer-controlled gas blender (RespirAct) was used to induce transient standardized isocapnic hypoxia in the lungs. A T2* weighted gradient-echo (GE) echo-planar imaging (EPI) sequence was used to acquire the imaging under controlled oxygen modulation. A custom Matlab script and SPM were used for preprocessing and analysis of the images. The blood oxygenation level-dependent (BOLD) response of tumor regions was assessed and compared to standard contrast-enhanced (CE)-T1 and FLAIR sequences. The initial tumor board decision was extracted for each case of radiographically suspected progression. It was categorized into “progression/recurrence” and “pseudoprogression/treatment related changes”. Six patients with radiographically suspected progression were included. Initial multidisciplinary discussion classified four as “progression” and two as “pseudoprogression”. In the progression group, contrast-enhancing regions showed consistent BOLD signal decrease in response to hypoxia. In pseudoprogression, larger areas of non-responsive tissue were observed. These preliminary findings suggest distinct BOLD signal patterns between the two entities. This pilot analysis suggests that hypoxia-modulated BOLD MRI may differentiate progression from treatment-related changes based on differential response to hypoxic modulation during BOLD imaging underlying different hemodynamic and oxygenation features. While the sample is small, this approach builds on earlier CO₂-based BOLD imaging studies in radiation necrosis, though the underlying physiological mechanisms are different.[3] 18F-(fluoroethyl)-l-tyrosine positron emission tomography (FET-PET) is the current gold standard to detect progression.[4] Ongoing recruitment and planned correlation with FET-PET imaging and clinical follow-up will allow for more definitive assessment of diagnostic utility. The combination of hypoxia-targeted BOLD-MRI with PET and perfusion techniques bears potential to further complement radiological follow-up in treated glioblastoma patients. The included patients will be longitudinally followed to determine which lesions prove to be true progression. Our initial observations indicate that hypoxia-targeted BOLD MRI may constitute an additional imaging biomarker for distinguishing true progression from pseudoprogression/radionecrosis in a multimodal imaging setting. Additional studies are necessary to evaluate BOLD responses in pseudoprogression cases. This strategy may enhance recurrence evaluation, resulting in more accurate future imaging of gliomas.
Tristan SCHMIDLECHNER (Zurich, Switzerland), Natalia CANTAVELLA FRANCH, Vittorio STUMPO, Jacopo BELLOMO, Christiaan Hendrik Bas VAN NIFTRIK, Martina SEBÖK, Michael WELLER, Andrea BINK, Zsolt KULCSAR, Luca REGLI, Jorn FIERSTRA
14:00 - 15:30
#47299 - PG403 Conventional MR spectroscopy sequences combined with machine learning allow distinguishing between IDH mutation and grade in astrocytomas.
PG403 Conventional MR spectroscopy sequences combined with machine learning allow distinguishing between IDH mutation and grade in astrocytomas.
Accurate grading and identification of IDH status in astrocytomas are essential for guiding therapeutic strategies and predicting patient prognosis. Their 2021 WHO classification emphasizes the importance of molecular markers, particularly IDH status, in tumor characterization [1]. Considering this, further investigation is needed to explore non-invasive diagnostic approaches. On the other hand, since 2012 when specific sequences were developed to detect the 2-hydroxyglutarate metabolite [2,3] conventional MRS sequences are not regarded as an option to detect the IDH mutation in gliomas. This study aims to re-evaluate the potential of magnetic resonance spectroscopy (MRS) in determining IDH status (mt: IDH mutated, wt: IDH wild type) and tumor grade (2, 3, or 4) by analyzing metabolite patterns across different astrocytoma subtypes, under the assumption that the effects of mutation and grade on the metabolic profile will affect enough the whole spectral pattern to allow for non-invasive discrimination.
The study utilized Single Voxel (SV) MRS data collected at Hospital de Bellvitge with 1.5T Philips Ingenia and Intera scanners in Spain. Five classification tasks were performed on Short Echo (SE, 30 ms), Long Echo (LE, 136 ms), and concatenated SE+LE spectra. Sequential forward feature selection and linear discriminant analysis were used to identify features to distinguish between: 1/Astrocitoma-IDH-mt-grade-2 (A2-mt) from Astrocitoma-IDH-NEC-grade-2 (A2-wt), 2/Astrocitoma-IDH-mt-grade-3 (A3-mt) from Astrocitoma-IDH-NEC-grade-3 (A3-wt), 3/A2+A3-wt from A2+A3+ Astrocitoma-IDH-mt-grade-4 (A4-mt), 4/A2 from A3, and 5/ A2+3 from A4.Classifiers were evaluated with area under the curve (AUC) and Balanced Error Rate (BER). The best classifier was defined by the smallest BER in the training phase. After application of inclusion criteria, there were 71 cases with SV MRS available for analysis at SE (15 A2-mt, 12 A2-wt, 20 A3-mt, 13 A3-wt and 11 A4-mt) and 69 cases at LE (14 A2-mt, 13 A2-wt, 18 A3-mt, 13 A3-wt and 11 A4-mt). Recurring discriminative spectral features include creatine, choline and lipids at SE and lactate and Glx, at LE which can be seen on the mean spectra (not shown). ). For the first question SE, and for the other questions the combined echo time, yielded the best classifiers. Notably, all AUC values for the best echo are quite high 0.938, 0.933, 0.834, 0.849 and 0.870 for the 5 questions, respectively. Histologically verified low-grade gliomas can be identified non-invasively, prior to surgery, with a conventional magnetic resonance imaging exam, due to their anatomical and contrast uptake characteristics. However, it is not as easy to distinguish between grades or IDH status. For this reason, a biopsy is needed, being performed before or during the removal of the tumour, in order to characterise the particular type of glioma.The discovery of molecular biomarkers of favourable prognosis in glioma subtypes, such as the IDH mutation was taken into account in the 2021 revision of the WHO classification of brain tumors [4]. These findings support our claim that IDH status can be successfully determined for A2 and A3, while A2 can be differentiated from higher grades of astrocytoma without needing specialized sequences [5].
Lili TOTH (Spain, Spain), Carles MAJÓS, Albert PONS-ESCODA, Carles ARÚS, Margarida JULIÀ-SAPÉ
14:00 - 15:30
#47396 - PG404 Stereotactic radiosurgery-induced changes in brain metastasis patients detected within two weeks after therapy using multi-parametric qMRI.
PG404 Stereotactic radiosurgery-induced changes in brain metastasis patients detected within two weeks after therapy using multi-parametric qMRI.
Brain metastases (BM) represent the most common malignant brain tumors in adults[1]. To obtain local control, neurosurgery and stereotactic radiosurgery (SRS) are competitive as well as complementary approaches that are further combined with systemic therapy[2]. Preoperative SRS offers the advantage of more precise target volume definition and reduces the risk of tumor spread into the cerebral spinal fluid space at the time of surgery[3]. Several MRI approaches have been taken to assess BM treatment efficacy, including synthetic MRI, quantitative MRI (qMRI), radiomics, magnetic resonance spectroscopy (MRS), and perfusion imaging[4–10]. With regard to SRS, treatment response is generally evaluated weeks after end of the treatment course[11]. For preoperative SRS, the availability of robust Quantitative Imaging Biomarkers (QIB) before surgery would be beneficial to assess treatment outcome and possible complications such as radiation necrosis on an individual patient basis. In this explorative study, we focus on early changes in conventional MRI contrast, which can be established from difference images calculated from pre-SRS and post-SRS MRI data. Image contrast in qualitative MRI-data (weighted-images) might be affected by hardware calibration, which may cause a bias in the difference images. This can be avoided using a quantitative MRI protocol (qMRI). Therefore, we used both, difference images from weighted MRI as well as qMRI to evaluate changes in BM following pre-operative SRS. In addition, we explored whether changes in automatic tumor segmentation mask volumes are consistent with the changes observed in the qMRI difference maps.
In vivo measurements were performed on six BM patients in two sessions (pre-SRS, post-SRS). The pre-SRS session was performed for SRS planning. The post-SRS session was performed within two weeks after SRS but prior to the surgical excision of symptomatic metastases. The measurements were carried out using a whole-body 3T MR Scanner (MAGNETOM Prisma (VE11C) or MAGNETOM Skyra (VE11C), Siemens Healthineers, Erlangen, Germany). In addition to the standardized Brain Tumor Imaging protocol (BTIP) [12], qMRI measurements were performed with a recently published vendor-based protocol[13]. Compared to the published protocol, the resolution was increased providing PD, T1, T2*, and QSM maps at 1mm isotropic. The planned radiation dose profile was later registered to the qMRI image space using FSL[14]. The BraTS-Toolkit was used to segment edema (OE), necrotic/cystic region (NE), and enhancing tumor (CE) masks. qMRI difference maps were computed by co-registering pre- and post-SRS qMRI maps, and calculating voxel-wise differences. For the regions WM, GM, NE, OE and CE, correlations were computed between the mean dose and the qMRI differences (post-SRS – pre-SRS). Fig. 1 shows an overview of the study and obtained MR data. CE mask volumes increased for the majority of patients. With regard to OE mask volumes, 50% decreased and 50% increased, independent of glucocorticoid dosage between pre- and post-SRS MRI. These volume changes corresponded to the changes visible in the qMRI difference maps. qMRI changes did not significantly correlate with radiation dose. Figure 2 demonstrates differing changes in tumor segmentation volumes for two patients. As seen in Figure 3, changes in OE differ between patients, with some showing a decrease in edema and some showing an increase after SRS. Figure 4 demonstrates the group level box-plots of the mean qMRI difference values for the different tissue classes. Paired t-tests revealed no significant differences between WM+GM and NE, CE and OE classes. While differences between normalized weighted-images revealed similar changes as for qMRI data for some subjects, it is not always possible to correct for all the transmitter and receiver field variations in the pre-RT and post-RT scans. qMRI data is preferred as QIB, as it ideally is independent of scanner- and session-related discrepancies. BRATs segmentations revealed variable changes in NE, CE and OE volumes across patients. These volume changes also correspond to the changes visible in the qMRI difference maps. The qMRI changes did not significantly correlate with radiation dose or with the cortisone-therapy dose, which implies that the observable change in the parameters is probably not just dose-dependent or cortisone-therapy dependent, but might signify individual tumor response to SRS. The different effects of SRS across patients may have predictive value for treatment outcome and possible complications such as radiation necrosis. These results demonstrate that changes in qualitative and quantitative MR scans of BM patients are detectable even within two weeks after SRS.
Dennis C. THOMAS (Frankfurt am Main, Germany), Svenja KLINSING, Mariem GHAZOUANI, Anna-Luisa LUGER, Seyma ALCICEK, Andrei MANZHURTSEV, Robert WOLFF, Marcus CZABANKA, Joachim P. STEINBACH, Ulrich PILATUS, Elke HATTINGEN, Pia S. ZEINER, Katharina J. WENGER
14:00 - 15:30
#47659 - PG405 Lactate, glutathione and GABA measurements in metastasis using MEGA-sLASER at 3T.
PG405 Lactate, glutathione and GABA measurements in metastasis using MEGA-sLASER at 3T.
Brain metastases (BM) are the most common malignant brain tumors in adults [1]. The therapeutic response to immune and targeted therapies remains unpredictable, however, reliable biomarkers could improve predictive accuracy. Recent advances highlight cerebrospinal fluid (CSF) profiling as a minimally invasive method to gain insights into the tumor microenvironment. Non-invasive metabolic imaging using MRS offers the potential to locally examine the tumor metabolism in vivo. In this study, edited MRS combined with qMRI is used to accurately measure absolute concentrations of lactate (Lac) not contaminated by lipids, glutathione (GSH) and gamma-aminobutyric acid (GABA) in BM. To our knowledge, for the first time the relationship between MRS-detectable Lac concentrations in BM and the Lac concentrations in CSF are analyzed.
Participants: 12 patients with BM: 4 with non-small-cell lung cancer, 3 with breast cancer, 2 with clear renal cell carcinoma, 2 with small-cell lung cancer and 1 with melanoma. Data were acquired using a 20-channel phased-array head coil on a 3T Siemens Prisma. Diagnostic imaging (3D T1w MPRAGE, 2D T2w, 3D FLAIR, 12 min) was performed, followed by MRS (15 min) and qMRI (8 min). Lumbar puncture was performed within 24 hours of MRI.
MRS was acquired in the BM area (Fig. 1A) and in the contralateral normal appearing brain tissue (CL) (Fig. 1B). For MRS, MEGA-sLASER pulse sequence [2] was used with TR = 2 s and TE = 80 ms. The 90 Hz frequency selective editing pulses were applied at frequencies δLac/GSH = 4.56 ppm, δGABA = 1.9 ppm, δOFF = 7.5 ppm to acquire 3x64 ONGABA, ONGSH/Lac, and OFFAll acquisitions, respectively. 3 unsuppressed water spectra were also acquired. Voxels of 20x20x20 mm3 were used in 8 patients, while 25x25x25 mm3 voxels were used in 4 patients with larger BM. For the qMRI, a vendor-based protocol (Thomas et. al [3]) was used to obtain PD and T1 maps. T2 maps were estimated from the T2w: 1) T2w were corrected for receiver field inhomogeneities in SPM [4]; 2) from the corrected T2w and PD map, the PDw was synthesized (sPDw) considering the mean WM T2 = 80 ms [5]; 3) T2 map was computed:
T2map = -TE/log(T2w_biascorr/sPDw)
MRS preprocessing was performed in Gannet [6] (MATLAB). Fitting and water-referenced quantification were performed in LCModel [7] using appropriate basis sets. Absolute concentrations were quantified using the Gasparovich et. al. method [8], water relaxation correction was performed using voxel-wise water concentration, T1 and T2 values calculated from the PD, T1 and T2 maps, respectively, using the in-house MATLAB code. For metabolite relaxation correction, literature T1 and T2 values were used. Considering that the tNAA signal originates from normal brain tissue only, a PVfactor was calculated accounting for the partial volume (PV) of the BM to determine the BM’s Lac concentration.
PVfactor = (tNAACL – tNAABM)/tNAACL Examples of the spectra acquired in one patient are shown in figure 2. For the BM, mean (SD) of the Cr SNR and the linewidth (LW) were 18.9 (5.9) and 5.1 (1.4) Hz, respectively. For the CL, SNR was 25.2 (9.5) and LW was 5.3 (1.4) Hz. High consistency was observed between the CL spectra. The water suppression factor was always >99%. PV-corrected Lac concentration was 7.5 (8.2) mM. Figure 3A shows the fitting quality of the Lac signal (without macromolecules), which was significantly better in the BM compared to CL. No correlation was observed between the MRS Lac levels and the CSF Lac level (figure 3B). The GABA and GSH concentrations were lower in metastasis compared to the CL (figure 4). In this study, MEGA-sLASER pulse sequence allowed to reduce lipid contamination of the Lac signal, providing high-quality spectra for Lac measurements. Furthermore, it allowed to get GABA, and GSH concentrations in BM, even with voxel sizes as small as 8 ml. The use of qMRI provided water relaxation parameters, which significantly impact metabolite quantification in lesions [9], including BM.
The absence of a correlation between MRS-derived BM Lac and lumbar-puncture derived CSF Lac could be due to the different sources of these parameters. MRS lactate reflects localized intra- or peritumoral metabolism, indicating hypoxia, altered glycolysis, and inflammatory changes [10]. In contrast, CSF Lac may remain near-normal even in the presence of highly glycolytic parenchymal BM, especially if there is no leptomeningeal involvement. The reduced concentrations of GABA and GSH in the BM regions most probably indicate the loss of normal cerebral tissue. Data acquisition and processing are ongoing. To improve corrections for the PV effects by more accurately estimating the tissue type fractions within the voxel, BraTS [11] will be performed. MEGA-sLASER combined with qMRI enables reliable Lac measurement in brain metastases. Voxel size limitations require partial volume effects correction. Yet, no correlation with CSF Lac was observed highlighting distinct metabolic sources.
Andrei MANZHURTSEV (Frankfurt/Main, Germany), Svenja KLINSING, Seyma ALCICEK, Dennis C. THOMAS, Anna-Luisa LUGER, Ulrich PILATUS, Katharina J. WEBER, Joachim P. STEINBACH, Elke HATTINGEN, Pia S. ZEINER, Katharina J. WENGER
14:00 - 15:30
#47852 - PG406 Evaluation of quantitative serial MRI of glioblastoma patients treated with immunotherapy.
PG406 Evaluation of quantitative serial MRI of glioblastoma patients treated with immunotherapy.
The use of immunotherapy in patients with brain tumours has been challenging. The immunotherapy response assessment for neuro-oncology (iRANO) criteria [1] were proposed to address challenges of immunotherapy, but require multiple visits, and often fail to distinguish between progression, pseudoprogression and treatment related enhancement.
This work aimed to evaluate serial quantitative MRI biomarkers [2] in patients with glioblastoma (GBM) who received immunotherapy in order to characterise early treatment response.
Eight GBM patients who received immunotherapy on a phase one clinical trial were assessed retrospectively. Each patient had an MRI examination at baseline and multiple post-treatment visits (range 2-6, depending on their evolution whilst on treatment). Cohort demographics are presented in Figure 1. MRI data were acquired between September 2019 and April 2024 using a clinical standard brain protocol including pre- and post-contrast T1w and diffusion-weighted imaging (DWI). Two scanners were employed in the study (1.5T Siemens, 6/8 patients for all of their visits; 3T Philips, 1/8 patients for all of their visits; one patient moved between scanners due to a system upgrade).
Three volumes of interest (VOIs) were delineated on the POST-contrast T1w and DWI acquisitions (Figure 1): VOI 1 and VOI 2 delineated the tumoral volumes on T1w and Apparent Diffusion Coefficient (ADC) images, respectively; VOI 3 (drawn on ADC map) included tumour and any surrounding abnormal tissue (e.g. oedema). All delineation was performed in Osirix [3]. The T1w resolution (1x1x1 mm3) was resampled to DWI (1.8x1.8x5 mm3), to allow copying of VOI 1 drawn on POST-contrast T1w data to ADC map. Enhancement fraction (EF) maps were calculated: EF=(POST-PRE)/ (POST+PRE). Median values of EF and ADC were reported for each patient at each visit; volumes of each type of delineation were also recorded. Histograms were used to demonstrate serial changes of EF and ADC.
Response was evaluated using iRANO criteria. Figure 1 depicts the baseline images for all patients (ADC, PRE and POST T1w) demonstrating the tumour heterogeneity of the cohort. Two patients (4 and 7) started the treatment after surgery. A common characteristic of the remaining 6/8 patients is the extended oedema tissue surrounding the tumour (Figure 1). Best response for iRANO is shown for all patients.
Overall, 5/8 patients demonstrated a rapid increase of tumoral volume (Figure 4) suggesting no response to treatment. These patients (3 to 7) stayed on treatment for shorter periods (58-201 days) than the responding patients (425-679 days).
Large changes of serial ADCs of tumour (VOI2) were experienced by three patients (2, 7 and 8) (Figures 2 and 4 ). Patient 4 also experienced large changes of ADC (Figure 4), but such changes were considered less reliable due to the reduced size of the tumour.
Patient 2 (orange line in Figure 4, CR by iRANO) showed a tumour ADC increase for several post-treatment visits (Figure 3) corroborated with stable tumoral volumes (Figure 4) suggesting a good response. At the same timepoints, EF has the largest increase suggesting non-response, which, based on the ADC changes, may be considered pseudo-progression.
Until visit 6, patient 8 (grey line in Figure 4, SD by iRANO) demonstrated increased ADC and stable EF and tumour volume, suggesting continuous good response. At visit 6, a large increase in tumour volume and an ADC decrease were observed, indicating non response. This change of treatment response is weakly suggested by the EF biomarker (showing a very small increase).
Patient 7 (pink line, PD by iRANO) demonstrated an example of a non-responder with large decrease in ADC and small increase in EF after treatment. (Figures 3 and 4).
Over the whole cohort, the serial ADCs of VOI3 (tumour+oedema) experienced smaller change than that of ADCs of VOI2 (tumour). This descriptive study is limited by the small number of patients, the tumour heterogeneity and the use of two scanners during the trial. Despite these limitations, the ADC biomarker was a better predictor of treatment response than EF in both responders and non-responders. The ADC biomarker was able to identify pseudoprogression for the first two post-treatment visits of patient 2, which was confirmed histologically. Moreover, the early increase in ADC observed for the best responder (patient 2) might suggest ADC as a promising biomarker for early treatment response assessment.
This study highlights the need to include quantitative MRI markers in treatment trials in order to prospectively evaluate which are the most informative. In this descriptive study, ADC biomarker showed promising results in characterising treatment response for patients with GBM above tumour volume and enhancement fraction alone and should be characterised further in a larger cohort of immunotherapy clinical trials.
Mihaela RATA (London, United Kingdom), Philip BENJAMIN, Matthew BLACKLEDGE, Diogo SILVA, Georgina HOPKINSON, Nina TUNARIU, Jessica WINFIELD, Juanita LOPEZ
14:00 - 15:30
#46951 - PG407 The Role of Diffusion-Weighted Imaging in Differentiating Pseudoprogression from Progression in Glioblastomas.
PG407 The Role of Diffusion-Weighted Imaging in Differentiating Pseudoprogression from Progression in Glioblastomas.
Glioblastoma (GBM) is the most common and aggressive primary brain tumor in adults, with a survival rate of 9–18 months [1]. Following treatment, patients may present with either true tumor progression (TP) or pseudoprogression (PsP), a treatment-related effect. Differentiating between TP and PsP is critical for appropriate clinical decision-making[1–3]. While conventional magnetic resonance imaging (MRI) plays a crucial role in brain tumor assessment, it is limited in distinguishing TP from PsP based solely on morphological imaging. Advanced techniques, such as diffusion-weighted imaging (DWI), offer additional information that may aid in this differentiation[2, 4, 5].
This retrospective study evaluated 49 patients diagnosed with GBM who underwent therapy and follow-up MRI scans. On apparent diffusion coefficient (ADC) maps, three regions of interest (ROIs) were drawn on the largest lesion area visible on contrast-enhanced T1-weighted images, and three ROIs were placed in the contralateral normal-appearing white matter. From each ROI, ADC mean (ADCmean), maximum (ADCmax), and minimum (ADCmin) values were extracted. Subsequently, ratios of ADC values between lesion and normal tissue (rADCmean, rADCmax, rADCmin) were calculated. Patients with TP showed lower ADC values compared to those with PsP. ADCmean, ADCmin, ADCmax, rADCmean, and rADCmax values were significantly different between the TP and PsP groups, while rADCmin did not show significant differences. White matter ADC values were significantly lower than lesion values but did not differ between TP and PsP groups. No significant differences were observed among the three lesion ROIs for ADC metrics within each group. In the white matter, all ADC metrics for the TP group and ADCmax and ADCmean for the PsP group were consistent across ROIs, except for ADCmin in PsP patients. ADCmax (AUC = 88.9%) and rADCmax emerged as the best metrics for distinguishing TP from PsP, with respective cut-off values of 1.822 × 10⁻³ mm²/s and 2.141. Both metrics achieved 83.3% sensitivity, 100% specificity, and 85.4% accuracy. Our findings highlight that DWI-derived ADC metrics, particularly ADCmax and rADCmax, are valuable biomarkers for differentiating TP from PsP. The significant differences in ADC values reflect underlying pathophysiological variations between true tumor growth and treatment-related changes[2, 4–6]. The high specificity and diagnostic accuracy demonstrated by ADCmax and rADCmax underscore the clinical potential of incorporating DWI analysis into routine GBM follow-up imaging protocols. DWI provides significant added value in the post-therapy differentiation between TP and PsP in GBM patients. ADC metrics, especially ADCmax and rADCmax, show strong diagnostic performance and can support more accurate clinical decision-making.
Freitas DAVIDE (Porto, Portugal), Nuno ADUBEIRO, Luísa NOGUEIRA, Inês OLÍMPIO
14:00 - 15:30
#47824 - PG408 Multiscale Tumor Characterization Using Diffusion MRI at 7T and 11.7T.
PG408 Multiscale Tumor Characterization Using Diffusion MRI at 7T and 11.7T.
Ultra-high-field magnetic resonance imaging (MRI) provides enhanced spatial resolution and tissue contrast, offering new opportunities for detailed tumor characterization [1]. However, current clinical MRI techniques often fail to reflect the heterogeneous nature of tumor tissue, limiting microstructural assessment [2]. Frequency-dependent multidimensional diffusion-relaxation correlation MRI (ωMDR-MRI) offers a novel framework to resolve sub-voxel features and characterize complex cellular environments. Recent nonparametric metrics further enable consistent assessment of tumor regions across scans and field strengths [3]. This study utilizes ωMDR-MRI at high resolution to improve detection of tissue anisotropy and microstructural features in gliomas, aiming to enhance both surgical precision and broader treatment planning.
A 74-year-old male glioma patient (WHO grade 4, IDH wildtype) was scanned preoperatively with a 1.5T Siemens MRI scanner. Whole tumor, contrast-enhancing tumor, and necrotic regions were segmented using 3DSlicer [4] from pre- and post-contrast T1-weighted MRI (TR = 2000 ms, TE = 2.69 ms, voxel matrix = 256 × 256 × 176, voxel size = 0.9766 × 0.9766 × 1 mm³), as shown in Figure 1A. Segmented voxels were z-score normalized, and histogram properties were analyzed for comparison. For visualization, the patient received Gliolan preoperatively. Following resection (Figure 1B), ex vivo scans were acquired at 7T and 11.7T using Bruker MRI systems, with parameters in Table 1.
Among the nonparametric maps derivable from ωMDR-MRI, squared normalized anisotropy was used due to its normalized formulation, enabling robust comparison across field strengths. It was calculated as the squared difference between parallel and perpendicular diffusivities, normalized by their sum, quantifying microscopic diffusion anisotropy from the frequency-dependent diffusion tensor. Five anatomically relevant ROIs were manually defined on ex vivo T1-weighted images: supramarginal zone of white matter (red), supramarginal zone of gray matter (green), tumor border (blue), and tumor regions (purple and yellow). Each ROI was co-registered via affine transformation (ANTs [5]) onto the anisotropy maps from both 7T and 11.7T (Figure 1C). Voxel-level comparisons between field strengths were performed using Wilcoxon signed-rank test with Dunn’s posthoc test. Within-field ROI comparisons were made using the Friedman test followed by pairwise Wilcoxon tests. Kernel density estimation (KDE) visualized signal distributions to highlight differences in central tendency and shape. Signal intensities from preoperative T1-weighted MRI showed statistical differences across tumor regions (Table 2A). Enhancing tumor areas had the highest mean intensities, followed by whole tumor and necrosis. The whole tumor showed the highest skewness, suggesting heterogeneity, while enhancing and necrotic areas showed lower skewness, indicating more balanced distributions. Pixel intensities in white matter, gray matter, and tumor regions differed significantly between 7T and 11.7T (p<0.01) and within each field strength (p<0.01) (Table 2B). No significant difference was observed between the tumor border and tumor regions at 7T; however, at 11.7T, the border region had significantly lower anisotropy (p<0.01). Signal intensities generally decreased at 11.7T (Figure 2). In white matter, distributions became more symmetric and less flat, suggesting improved homogeneity. Gray matter showed increased skewness and kurtosis, indicating higher variability. The tumor border showed flatter, more asymmetric distributions, suggesting structural heterogeneity. In tumor regions, increased positive skewness at 11.7T pointed to better detection of intratumoral complexity. We found increased homogeneity in white matter, higher signal variability in gray matter, and clearer structural heterogeneity in the tumor border region at 11.7T. These results highlight the improved sensitivity of ultra-high-field imaging to microstructural features. The tumor region’s positively skewed distribution supports better detection of intratumoral complexity at higher field strengths. Additionally, while 7T did not differentiate between the border and tumor regions in terms of anisotropy, 11.7T revealed significantly lower anisotropy in the border zone, suggesting increased sensitivity to peritumoral heterogeneity. These findings emphasize the potential of ultra-high-field imaging to enhance tissue characterization in challenging regions. Future steps of this multiscale study will include microscopy image integration to assess cellular and subcellular structures, offering a refined understanding of the tumor microenvironments. This study demonstrates that ultra-high-field MRI at 11.7T provides improved sensitivity to tumor tissue microstructure and may support more accurate diagnosis and treatment planning in gliomas.
Buse BUZ-YALUG (Espoo, Finland), Omar NARVAEZ, Minna NIITTYKOSKI, Ville LEINONEN, Susanna RANTALA, Jussi TOHKA, Alejandra SIERRA
14:00 - 15:30
#46035 - PG409 Probing tumor microstructure in soft-tissue sarcomas using quantitative MRI combined with image-guided biopsy and digital pathology.
PG409 Probing tumor microstructure in soft-tissue sarcomas using quantitative MRI combined with image-guided biopsy and digital pathology.
Soft-tissue sarcomas (STS) are rare tumors with significant inter- and intra-tumor heterogeneity. Consequently, there is a need for imaging methods to guide biopsy[1], targeting heterogeneous components, and for evaluation of novel therapies. MRI enables quantitative assessment of tumors but better understanding of the link between quantitative MRI and tumor microstructure is required. Developments in image-guided biopsy[2] provide an opportunity to target biopsy sites using robotic guidance combined with quantitative MRI, while advances in digital pathology including multiplex immune-fluorescence (mIF) enable rapid characterization and quantification of cells in biopsy samples[3]; combining these techniques provides an opportunity to evaluate quantitative MRI parameters and corresponding tumor microstructure. The aims of this study are (a) develop a workflow combining image-guided biopsy with digital pathology in STS; (b) use this methodology to evaluate correlation between apparent diffusion coefficient (ADC) and numbers of cells, and between enhancement fraction (EF) and endothelial cells.
Patients were recruited at one centre as part of a prospective study[2] with written consent for use of tissue samples.
Patients underwent an MRI examination before biopsy (1.5T MAGNETOM Sola, Siemens Healthineers, Forchheim, Germany) described in Fig.1.
Antibodies were optimized for immunohistochemistry (IHC), multiplex positions and validated with single-plex IF (Opal TSA system, Akoya). Controls were used for IHC and IF optimizations. Two panels of mIF were optimized. Panel A was used to identify tumor cells, T-cells and B-cells. Panel B was used to identify macrophages, endothelial cells and cancer-associated fibroblasts (CAFs). Panels are described in Fig.2. Whole-slide images were generated using a Leica Bond autostainer and mIF images were acquired using a PhenoImagerHT (Akoya). Image analysis was done using HALO image software (IndincaLabs). Panels A and B were analysed for each biopsy sample to output the total number of cells and number of tumor cells, tumor-infiltrating lymphocytes (TILs=T-cells+B-cells), macrophages, endothelial cells and CAFs per unit area.
Correlation between cell types was assessed using Pearson correlation coefficient. Correlation between ADC or EF and number of cells per unit area was assessed initially using Pearson correlation coefficients across all samples. Further analysis used a linear mixed effects model to evaluate the effect of each cell type on ADC, with number of cells per unit area as fixed effects (after log transformation and normalization) and patients as a random effect (fitlme, Matlab2021a). The variance partition coefficient (VPC) was used to assess inter-patient variation in ADC[4]. 32 biopsy samples from 11 patients (2-3 samples per patient) were included.
Median ADC across all samples was 1570x10-6mm2/s (range (656-2830)x10-6mm2/s). Median/range of each cell type are shown in Fig.3.
Correlation between number of each cell type per unit area was weak-moderate (r=0.2-0.6) except between macrophages and CAFs, which were strongly correlated (r=0.7).
ADC showed negative correlation with total number of cells per unit area and negative correlation with number of tumor cells, macrophages and CAFs per unit area; correlation between ADC and TILs or endothelial cells was weak (Fig.3).
Mixed effects modelling included tumor cells, macrophages and CAFs (excluding TILs and endothelial cells). Number of CAFs per unit area had a significant effect on ADC (p=0.01) but numbers of macrophages and tumor cells did not have significant effects (p=0.7, p=0.9). VPC was <0.1.
No correlation was observed between EF and number of endothelial cells per unit area (Fig.4). This study demonstrated a workflow combining quantitative MRI, image-guided biopsy and digital pathology for validation of imaging biomarkers and investigation of tumor heterogeneity.
Correlation between ADC and total number of cells per unit area confirms that ADC is inversely related to cellularity in STS. However, the significant effect of CAFs on ADC in the mixed effects model shows that interpretation of ADC estimates depends on tumor stroma, not solely on tumor cells. The small VPC suggests inter-patient variation is small compared with total variation across all samples.
The absence of correlation between EF and endothelial cells is consistent with previous studies[5] suggesting that enhancement depends on the function of the tumor vasculature rather than presence of vessels.
A limitation of the study was the small sample size. It was not possible to analyse subtypes separately as 9/11 tumors were liposarcomas; future studies will investigate other subtypes. ADC is correlated with total number of cells per unit area but the number of CAFs per unit area is also significant indicating the importance of stroma in STS. This study demonstrates a workflow combining quantitative MRI and image-guided biopsy with digital pathology in STS.
Jessica M WINFIELD (London, United Kingdom), Edward W JOHNSTON, Amani ARTHUR, Geoff CHARLES-EDWARDS, Paul HUANG, Robin L JONES, Manuel SALTO-TELLEZ, Khin THWAY, Brandon WHITCHER, Tom LUND, Christina MESSIOU
14:00 - 15:30
#47053 - PG410 Multi-regional, automatic and volumetric temperature regulation during in vivo MRI-guided laser-induced thermotherapy (MRg-LITT).
PG410 Multi-regional, automatic and volumetric temperature regulation during in vivo MRI-guided laser-induced thermotherapy (MRg-LITT).
Mini-invasive MR-guided Laser Interstitial Thermal Therapy (LITT) is used to treat tumors in various organs (brain [1], liver [2], prostate [3]) using one or more optical fibers [4–6]. However, clinical procedures often rely on predetermined laser power and exposure time, which can lead to the target area being over- or under-heated. A multi-probe laser device is presented, whose output power is automatically regulated in real-time from MR-temperature maps to force tissue temperature to follow a predefined desired temperature-time profile.
LITT device:
A prototype multichannel LITT system (Alphanov, France) was connected to three independent optical fibers (400 µm diameter), each terminated by a diffuser tip (2 cm length, 1.8 mm diameter). Fibers were inserted into the leg muscle of an anaesthetized pig (~35 kg, ethics approved), forming a triangle with ~5 mm spacing. Each fiber was independently connected to a 976-nm laser diode that was controllable and interfaced with the thermometry pipeline [7].
MR-Imaging workflow:
The procedure was performed on a 1.5 T clinical scanner (Avanto, Siemens, Germany) using a 4-channel surface coil placed above the leg and two elements embedded into the MRI bed coil) for signal reception. The acquisition protocol included: (1) anatomical imaging for probe positioning, (2) real-time MR thermometry during treatment, and (3) post-ablation assessment.
Anatomical scans were acquired using a 3D MPRAGE (TI=1100 ms, TE=3.3 ms, TR=2000 ms, FA=15°, voxel size: 1.17×1.17×1 mm, FOV: 300×253×120 mm), before and after probe insertion into the muscle.
A stack of 10 slices was positioned over the probe tips to perform PRFS-based MR-thermometry [8]. Phase images were acquired at 1 Hz using a single-shot EPI [9] (TE=21 ms, TR=1000 ms, FA=50°, voxel size: 1.56×1.56×3 mm, GRAPPA=2, partial Fourier=6/8, bandwidth=1150 Hz, FOV: 200×200 mm). Thermal maps were computed in real time and displayed on the fly ( Certis Therapeutics software).
After completion of the thermal treatment, non-perfused volumes were visualized from 3D T1-weighted images (TE=2.16 ms, TR=4.49 ms, FA=10°, voxel size: 1.19×1.19×1.2 mm, FOV: 308×380×120 mm) acquired one minute after intravenous injection of gadoteric acid injection (0.5 mmol/kg).
Regulation algorithm:
The regulation algorithm was implemented in Gadgetron to automatically control heat deposition in real-time in several Regions of Interest (ROIs) centered on each laser. Input parameters: (1) the temperature-time profiles defined for (2) each ROIs (3x3x10 pixels each) positioned around each diffuser, (3) tissue thermal parameters (absorption and thermal diffusivity) determined from a calibration step, and (4) the heating pattern of each laser source. A PID controller combined with the Bio-Heat Transfer Equation was implemented [10]. At each acquired temperature stack [11], the power of each source was updated.
Calibration step:
Before temperature regulation, a constant power of 6 W was sequentially applied under MR-thermometry monitoring during 30 s on each diode, interleaved with a cooling period of 50 s, and the hottest voxel in temperature maps associated to each fiber was automatically detected and served as input for automatic ROI positioning. The temperature fitting method based on the BHTE equation was used to estimate the absorption and thermal diffusivity (D) [10], as well as the source function Si [11]. Figure 1 shows an example of 3D scan performed after inserting the laser probes into the muscle. The stack of slices for MR-thermometry is indicated by blue rectangles.
Figure 2 shows examples of automatic temperature regulation with (Top) an identical target heating profile applied to the 3 ROIs (+30°C, 275 s); and (Bottom) different heating profiles applied on 2 ROis: a single plateau (+25°C, 180 s) applied on the first ROI, and 2 plateaus of +15°C and +30°C on the second ROI, both lasting 60 s.
Table 1 summarizes metrics to evaluate the regulation algorithm performances. For both experiments and all ROIs, the mean ± std of the difference between target and measured temperatures remained below 0.3 ± 1.7 °C.
Figure 3 shows the temperature and thermal dose images for all slices, illustrating the possibility to modulate the size and shape of the treated volume. The MR-thermometry based automatic regulation is precise enough to force the temperature to follow predefined profiles during multi-probe LITT. This multi-region control of heat deposition enables conformable therapy and may prevent overheating of critical structures in order to increase MRI-controlled treatment efficiency and safety.
Manon DESCLIDES (Bordeaux), Valéry OZENNE, Pierre BOUR, Thibaut FALLER, Guillaume MACHINET, Christophe PIERRE, Julie CARCREFF, Stéphane CHEMOUNY, Bruno QUESSON
|
Poster hall |
15:30 |
TIME FOR A BREAK
- Coffee and refreshments will be available at the cash bar.
|
15:40 |
"Friday 10 October"
Added to your list of favorites
Deleted from your list of favorites
A25
15:40 - 17:10
FT2 Oral - Brain Connectivity, Structure, and Biomarkers
15:40 - 15:50
#45997 - PG022 Structural Brain Pathways Linking White Matter Hyperintensities to Pain Sensitivity in the Rotterdam Study.
PG022 Structural Brain Pathways Linking White Matter Hyperintensities to Pain Sensitivity in the Rotterdam Study.
Despite affecting one in five European adults and ranking as the top cause of disability [1], chronic pain (CP) severity often poorly correlates with tissue damage, pointing to central neuroplastic mechanisms. While prior studies have reported associations between structural brain changes—particularly in white matter—and pain [2], the causal pathway from white matter change to altered pain perception remains unclear. Here, we test whether white matter hyperintensities (WMH) alter pain sensitivity by disrupting tract microstructure and inducing cortical atrophy in connected regions (Figure 1).
We analyzed data from 1,447 participants (mean age 73 ± 6 years), in the Rotterdam Study, a large population-based cohort in the Netherlands. Pain sensitivity was assessed using the cold pressor test (CPT), where participants submerged their hand in 3°C water for as long as tolerable, up to a maximum of 120 seconds (Figure 2). This provided a quantitative measure of individual pain sensitivity.
Available brain MRI data included T1-weighted structural images, fluid-attenuated inversion recovery (FLAIR) sequences, and diffusion tensor imaging (DTI). Cortical volume was derived from the T1-weighted images using FreeSurfer. DTI data were preprocessed using the FSL pipeline [3] to derive fractional anisotropy (FA) and mean diffusivity (MD), and to generate a tractography-based template defining 27 major white matter tracts. We calculated mean FA and mean MD values within each predefined tract. Segmented WMH volumes, obtained using the PGS pipeline [4]—a trained deep learning-based approach—were then mapped onto the predefined tracts.
We conducted tract-wise regression analyses to examine associations between WMH burden, DTI metrics, cortical volume, and CPT performance. Mediation analyses were performed to identify indirect pathways linking WMH to altered pain sensitivity through DTI or cortical changes. Covariates included age, sex and intracranial volume. Among the 1,447 participants, 404 reached the maximum test duration of 120 seconds, while 1,043 withdrew their hand earlier. Among those who withdrew early, the mean CPT duration was 34.5 ± 23.0 seconds (Figure 2). WMH were detected in 20 out of the 27 predefined tracts.
Regression analysis showed higher WMH burden within all these 20 tracts was significantly associated with reduced FA values (Figure 3). Lower FA was further linked to greater pain sensitivity, particularly in the left anterior thalamic radiation (ATR_L) tract (multiple testing corrected p < 0.004) and the right medial lemniscus (ML_R) tract (multiple testing corrected p < 0.030) (Figure 4). In addition, higher WMH burden in the ATR_L tract was directly associated with higher pain sensitivity (multiple testing correction p<0.006) (Figure 4).
Mediation analysis confirmed that 78% of the effect of WMH in the ATR_L tract on pain perception was mediated by FA reduction in the same tract (average causal mediated effect (ACME) p < 2e-16), while the direct effect was not statistically significant (average direct effects (ADE) p = 0.630). Mediation analysis further supported a second pathway: WMH in the left corticospinal tract (CST_L) → cortical degeneration in the left postcentral gyrus → increased pain sensitivity, where 14% of the total effect mediated through cortical degeneration (ACME p<0.008), while the direct effect was not statistically significant (ADE p = 0.122). These results are the first to map tract-specific WMH impacts on pain sensitivity in a population cohort. Our findings suggest that WMH contribute to increased pain sensitivity through two mechanisms: disruption of white matter tracts and degeneration of cortical regions connected to these tracts. The ATR tract is known to be involved in cognitive, emotional and executive functions, whereas the CST is associated with sensory-motor integration and feedback. They appear particularly susceptible to WMH-related changes that alter pain processing.
Our findings are further supported by previous studies that have identified these tracts in the context of pain and sensory. For example, lower FA in the ATR tract has been reported in individuals with interstitial cystitis/bladder pain syndrome, a chronic pain condition [5]. In addition, the postcentral gyrus, which is connected to CST, is known to be directly involved in processing touch sensations [6]. These existing findings align with our observations of pain sensitivity associated with WMH burden in sensory pathways.
Together, our results support the notion that vascular pathology in white matter may influence central pain perception through tract-specific mechanisms, highlighting potential targets for future research and clinical intervention. Our findings contribute to a more detailed understanding of the neurobiological mechanisms underlying pain and offer a structural framework for future multi-cohort studies and collaborative meta-analytic investigations.
Xianjing LIU (ROTTERDAM, The Netherlands), Lieke KUIPER, Torgil VANGBERG, Meike VERNOOIJ, Christopher NIELSEN, Joyce VAN MEURS, Gennady ROSHCHUPKIN
15:50 - 16:00
#47958 - PG023 Toward direct Mapping of the Human Pallido-Subthalamic Pathway In Vivo at 3T.
PG023 Toward direct Mapping of the Human Pallido-Subthalamic Pathway In Vivo at 3T.
The subthalamic nucleus (STN) is a key integrative hub of basal ganglia circuits (Nambu 2011; Hamani 2004). However, the direct connection from the globus pallidus externus (GPe) to the STN – the pallido-subthalamic pathway – remains poorly characterized in humans. Ex vivo 11.7T diffusion MRI and histology have revealed a complex, topographically organized projection along this pathway, but mapping it in vivo at 3T is challenging due to lower spatial resolution and crossing fibers in the subthalamic region (Coenen 2022). Here, we leverage ex vivo findings to guide in vivo 3T tractography. We acquired diffusion MRI data from four healthy human subjects at 3T to test whether the ex vivo connectivity pattern can be reproduced in vivo. The ultimate goal is to enable direct in vivo mapping of the pallido-subthalamic tract, which could inform clinical interventions such as deep brain stimulation (DBS).
Four healthy volunteers were scanned on a 3T CIMA.X (Siemens Healthineers) equipped with ultra‑intense Gemini gradients (Gmax = 200 mT m⁻¹, slew = 200 T m⁻¹ s⁻¹) and a 64‑channel head coil (CENIR, Paris). The protocol combined a 3‑D T1‑weighted M2PRAGE and a multi‑shell diffusion acquisition using the CMRR multiband 2‑D EPI sequence (TE = 53 ms, TR = 2600 ms, multiband = 4, iPAT = 2, partial‑Fourier = 6/8, flip = 90°, EPI factor = 104). Diffusion sampling comprised 5 b0, 65b = 1500, 65 b = 3000, 40 b = 4000, 40 b = 6000 s mm⁻² voxel size = 1.5 mm³; this DWI sequence duration was 10 min 08s and it was repeated twice with a reversed phase encoded direction .
For comparison, a post‑mortem female brain (78 y) was immersion‑fixed in 4 % PFA (8 d, 4 °C) and imaged on an 11.7 T Bruker BioSpec (72 mm transceiver). A T2* (110 µm) and a multi‑shell diffusion scan (7 b = 1000, 29 b = 4000, 64 b = 10000 s mm⁻², 0.22 mm³; TR/TE = 250/25.2 ms; 61 h) were acquired.
All diffusion data underwent denoising, Gibbs‑ringing removal, distortion/motion/eddy correction (Topup & Eddy, FSL) and bias‑field correction; NORDIC‑PCA was additionally applied to the 3 T data (Moeller 2021). Multi‑shell multi‑tissue CSD produced fibre‑orientation densities. Probabilistic tractography (MRtrix3 iFOD2; FA > 0.07; length 1.1–25 mm) was seeded bidirectionally in the GPe; streamlines entering the internal capsule were excluded. Resulting tracts were classified into limbic, associative and sensorimotor subdivisions using a histology‑derived GPe atlas (Bardinet 2009) and mapped to their corresponding STN territories (Tournier 2019). In all four subjects, tractography delineated an ascending pallido-subthalamic tract from the GPe to the STN (bypassing the GPi), consistent with ex vivo anatomy. The STN projections were organized similarly in vivo and ex vivo: limbic fibers terminated in anterior STN, motor fibers in posterior-ventral STN, and associative fibers in central STN. The ex vivo data showed only a slightly broader sensorimotor termination area, attributable to its higher resolution. Fiber orientation density (FOD) analysis confirmed pronounced crossing-fiber configurations where the pathway traverses the internal capsule. Multi-shell tractography resolved these crossings, enabling clear identification of the pathway within the dense internal capsule. The agreement between in vivo and ex vivo patterns across subjects demonstrates the feasibility of mapping this pathway in vivo at 3T. This study provides the first in vivo visualization of the human pallido-subthalamic pathway on a 3T MRI. Our tractography reproduced the limbic, associative, and sensorimotor segregation of GPe–STN fibers previously seen only ex vivo. These results show that advanced diffusion MRI at 3T can map small, complex subcortical tracts in vivo (Coenen 2022). The close correspondence between in vivo and ex vivo connectivity patterns indicates that the STN’s functional territories are preserved in vivo. Minor differences, such as a slightly larger sensorimotor territory ex vivo, likely reflect the ultra-high resolution and absence of motion in the postmortem data. By showing that a 3T scan can resolve this pathway, we lay groundwork for clinical translation. In neurosurgical planning, patient-specific tractography of the pallido-subthalamic pathway could enhance DBS targeting by revealing individual fiber anatomy. Future studies in patients (e.g., Parkinson’s disease) should examine how variations in this pathway relate to symptoms and treatment outcomes. We mapped the pallido-subthalamic tract in vivo at 3T, replicating the three-part (limbic, associative, motor) projection pattern observed ex vivo. Despite the lower resolution of in vivo imaging, the key features of this pathway were identifiable in all subjects. This demonstrates that high-quality diffusion MRI at 3T can visualize detailed subcortical connections in living humans, potentially improving precision in neurosurgical targeting and therapy for basal ganglia disorders.
Nicolas TEMPIER (Paris), Mélanie DIDIER, Christophe DESTRIEUX, Mathieu SANTIN, Chantal FRANÇOIS, Carine KARACHI, Eric BARDINET
16:00 - 16:10
#46451 - PG024 Detailed Investigation of the IhMT Anisotropy in Ex Vivo Spinal Cord Under Sample Reorientation.
PG024 Detailed Investigation of the IhMT Anisotropy in Ex Vivo Spinal Cord Under Sample Reorientation.
Magnetization transfer (MT) and inhomogeneous MT (ihMT) are sensitive to motion-restricted macromolecules associated with residual dipolar couplings (RDCs) and have been extensively exploited to probe myelin-lipid bilayers of white matter (WM) tracts [1,2]. RDCs between ‘non-aqueous’ protons in an approximately cylindrical arrangement, such as WM fibers, are intrinsically anisotropic and have the potential to significantly modulate (ih)MT contrast in in vivo MRI. In previous work, Pampel et al. were able to interpret the anisotropy of MT using a cylindrical fiber model [2]. Recent studies investigating the orientation dependence of ihMT in model systems [1], and in WM in vivo [3-5] and ex vivo [6,7] have yielded conflicting observations. IhMT anisotropy effects have been shown to be dependent on offset frequencies (ΔRF) in ex vivo spinal cord, consistent with changes in the cylindrical absorption lineshape [6,7]. A full explanation of anisotropy effects remains elusive, given the multiple potentially orientation-dependent parameters, their mutual correlations, and the scope of experimental datasets required for detailed analysis. To address this research gap, measurements were performed on ex vivo spinal cord with a randomised variation of the fiber-to-field angle (θFB) to reveal the 'true' anisotropy of ihMT.
A white matter section was extracted from fixated porcine spinal cord (post-mortem interval of 2-3 hours), washed in PBS, and placed in a 5-mm NMR tube (filled with Fomblin). MR measurements were conducted at 3T (MAGNETOM Sykrafit) using a custom-made TxRx Helmholtz coil at ~35-36°C [8]. Acquisitions were performed with a 1D CPMG readout (0.483 mm nominal resolution, 80 echoes, echo-spacing 4 ms, 200 μs composite refocusing pulse) following a rectangular pulse (20 μs), allowing for future planned ‘four-pool’ model analyses [9, 10]. Here, the focus is limited to the intensity of the first echo as well as on the ihMT-related experiments (acquired after a train of 2-ms Gaussian pulses, NRF=300), including:
(I) ‘ihMT sets’: no MT preparation (MToff), single-sided (MT+, MT–) and dual-sided irradiation with alternating offset (MTalt) or cosine-modulated pulse (MTcos) for different offsets (ΔRF),
(II) z-spectra acquisitions including (MT+, MT–, MTalt, MTcos).
A central ROI (11 voxels) was selected for the analysis. By not accounting for the multi-exponential ‘T2-decay (due to the presence of myelin water and intra/extracellular water)’, the analysis can be restricted to a single ‘aqueous’ compartment - to a first approximation. A two-pool model (2PM) with a dipolar reservoir is used to keep the simultaneous fit (using custom routines [2,11,12]) of the z-spectra (for ΔRF >2 kHz, 1716 data points for 10+1 orientations) as simple as possible, focusing on the ability of the proposed ‘fiber model’ to explain the ihMT anisotropy. The model-free analysis of the z-spectra (Fig. 1) shows (indirectly) the orientation-dependent lineshape changes of the non-aqueous pool.
An unambiguous orientation dependence was observed for ihMT ratios, with remarkably strong orientation-dependent variations and strikingly different trends as a function of θFB observed for different ΔRF, e.g., for θFB= 0° max for ΔRF<15 kHz vs. min for ΔRF≥20 kHz (Fig. 2). Thus, statements about ‘higher ihMTR in fibers running parallel to B0’ [3,4] apply only to certain experimental conditions (e.g., ΔRF < 15 kHz here).
Further insight into the anisotropy of (ih)MT can be obtained by the degree of anisotropy for each offset ΔRF (Fig. 3). A novel finding is the ‘pattern’ with two maxima for ihMTR anisotropy (Fig. 3B). Model 1 used a set of 7 isotropic parameters with a fit of T2B as a function of θFB, equipped with a super-Lorentzian lineshape (Fig. 4A-C). The general trend of the experimentally observed ihMTR anisotropy could be reproduced, suggesting that most of the orientation dependence for (ih)MT is related to changes in the absorption lineshape although other spin system parameters may also show anisotropy [12]. In model 2, the number of fit parameters is reduced by more than half, while the orientation dependence of the lineshape is derived from the ‘cylinder model’ for a single axon, resulting in a qualitative agreement of ihMTR anisotropy with the experimental trend (Fig. 4D-F). The overestimated anisotropy effect of model 2 is somewhat to be expected, as fiber dispersion effects have not yet been considered. A strong dependence of ihMTR(θFB) on ΔRF could undoubtedly be confirmed in fixed spinal cord as a model for highly ordered WM tracts. The novel finding of a characteristic ‘pattern’ of ihMTR anisotropy with two distinct maxima for different offsets will provide a benchmark for future model evaluation. Initial results obtained from a simple 2PM with a ‘cylindrical lineshape’ are in qualitative agreement, indicating the potential to derive unbiased ihMT-related metrics when accounting for θFB, e.g. by combining ihMT and and DWI experiments.
Niklas WALLSTEIN (Marseille), André PAMPEL, Guillaume DUHAMEL, Roland MÜLLER, Carsten JÄGER, Harald E. MÖLLER, Olivier M. GIRARD
16:10 - 16:20
#46774 - PG025 Interpretable Machine Learning Model for Characterizing Magnetic Susceptibility-based Biomarkers in First Episode Psychosis.
PG025 Interpretable Machine Learning Model for Characterizing Magnetic Susceptibility-based Biomarkers in First Episode Psychosis.
Recent publications highlighted the changes in brain iron concentrations with a co-factor in dopamine pathways in psychosis patients (1). Insights about the quantification of iron concentrations in the brain are now available through effective transverse relaxation rate (R2*) and quantitative susceptibility mapping (QSM), calculated from multi-echo gradient-echo (GRE) sequences (2). As iron is the cofactor in neurotransmitter biosynthesis, the Grey Matter (GM) nuclei functions are susceptible to changes in iron concentration. Considering the limited number of dopamine pathways (nigrostriatal and tuberoinfundibular pathways) retrieved from the QSM image, it is possible to refine disease monitoring and improve patient risk stratification (3). The present study aims to pinpoint potential predictive biomarkers derived from QSM and R2* for individuals experiencing first-episode psychosis (FEP), along with their response to antipsychotic treatment.
3D multi-echo GRE and T1-weighted FLAIR of 52 healthy volunteers (HV) and 78 FEP patients (52 RS and 24 TRS) were acquired in a 3T Philips Ingenia MRI scanner. QSM reconstruction was performed as in (4) using Variable Sophisticated Harmonic Artifact Reduction for Phase data (vSHARP) (5) and FAst nonlinear Susceptibility Inversion (FANSI) toolbox (6). Images were registered and normalized to an NMI space. Twenty-two regions of interest (ROI) of deep GM and subcortical brain nuclei were segmented using the Multicontrast PD25 version 2019 (7). We calculated each ROI's mean QSM and R2* values, as shown in Figure 1. This study developed two machine learning models to analyze brain regions with QSM and R2* values, distinguishing between HV and FEP patients, responders (RS), and treatment-resistant (TRS) patients. The models were built using Python and employed RF with Sequential Forward Selection (SFS) for feature selection, hyperparameter optimization through grid search, and 10-fold cross-validation. SHAP values were used for model interpretation, revealing feature importance (8). The classification results for the HV vs. FEP classification (Figure 2), four features—right nucleus accumbens R2*, left amygdala R2*, left nucleus accumbens R2*, and right thalamus QSM—yielded an accuracy of 76.48 ± 10.73%. For RS vs. TRS, four features—right hippocampus R2*, left caudate R2*, left putamen R2*, and left amygdala QSM—achieved 76.43 ± 12.57% accuracy. After applying feature selection, treeSHAP analysis was conducted to identify the most important predictors in the model, shown in Figure 3. The SHAP summary plots showed that the key features for HV vs. FEP classification were right nucleus accumbens R2*, left amygdala R2*, left nucleus accumbens R2*, and right thalamus QSM. For RS vs. TRS, the important features were left amygdala QSM, right hippocampus R2*, left caudate R2*, and left putamen R2*. Both classifications use similar features, leading to confusion between classes. The SHAP values indicate the contribution of each feature to the model's predictions, with lower right nucleus accumbens R2* values increasing the likelihood of being classified as FEP, while higher left amygdala R2* values favor HV classification. Figure 4 presents the brain areas found as disease predictors by treeSHAP per map and classification problem. This study identified the most relevant QSM and R2* features for predicting First-Episode Psychosis (FEP) and treatment response using Random Forest (RF) models. Two classification problems were addressed: HV vs. FEP and RS vs. TRS. Feature selection reduced the number of input variables while maintaining model performance, achieving accuracies of 76.48% for HV vs. FEP and 76.43% for RS vs. TRS. SHAP analysis was used for global and local interpretability, revealing the top predictive features, including the right nucleus accumbens R2* and left amygdala QSM. The study emphasized the complementary nature of QSM and R2* in understanding tissue magnetic properties, particularly regarding brain iron content linked to schizophrenia. Despite some limitations, such as small sample sizes and the use of classical ML algorithms, the findings support the relevance of iron-sensitive imaging features in the context of psychosis. The study demonstrated the potential of QSM and R2* imaging biomarkers for early detection of treatment-resistant schizophrenia, which could help improve clinical outcomes. Future research will explore advanced deep-learning methods and larger datasets to enhance classification performance and model generalizability.
Pamela FRANCO, Cristian MONTALBA (Santiago, Chile), Raúl CAULIER-CISTERNA, Carlos MILOVIC, Alfonso GONZÁLEZ, Juan Pablo RAMIREZ-MAHALUF, Juan UNDURRAGA, Rodrigo SALAS, Nicolás CROSSLEY, Cristian TEJOS, Sergio URIBE
16:20 - 16:30
#47675 - PG026 Validation of fetal brain 3D slice-to-volume registration (SVR) in detecting the cause of antenatal ventriculomegaly confirmed by neonatal scan.
PG026 Validation of fetal brain 3D slice-to-volume registration (SVR) in detecting the cause of antenatal ventriculomegaly confirmed by neonatal scan.
Detecting subtle anatomical abnormalities in fetal brain MRI is particularly challenging due to motion artefacts and the limited spatial resolution of 2D slices[1]. Motion artefacts caused by fetal movement and maternal breathing can result in inconclusive or inaccurate antenatal diagnoses, leading to unnecessary stress to patients and termination of pregnancy[2].
To address these limitations, slice-to-volume registration (SVR) software has recently been developed. SVR aligns multiple 2D MRI slices into a single, high-resolution 3DSVR[3]. The reconstructed 3DSVR can be reoriented in any plane via multiplanar reconstruction (MPR), thus improving diagnostic accuracy and confidence[2]. To date, validation studies have focused mainly on comparing biometrical measurements and radiological assessment between 2D and 3DSVR, showing the superiority of 3DSVR[4,5]. However, there is a lack of studies confirming pathological findings on postnatal imaging and/or outcomes.
This study seeks to address this gap by investigating the effectiveness of 3DSVR, compared with 2D, for identifying anatomical abnormalities in fetal MRI and assessing its impact on image quality in a cohort of fetuses diagnosed with antenatal ventriculomegaly confirmed postnatally.
Subjects:
A cross-sectional study was conducted with 20 pregnant participants diagnosed antenatally with ventriculomegaly. All had both fetal and neonatal MRI scans available. Median gestational age(GA)at fetal MRI was 28w+2d (21w+1d/33w+3d); neonatal MRI was performed at median corrected GA of 38w+1d (32w+5d/42w).
Acquisition Protocols:
Fetal and neonatal MRI was acquired using standard clinical protocols. Fetal imaging was performed using 1.5T Siemens. All fetuses underwent T2-weighted HASTE (half Fourier single-shot fast spin echo) with the following parameters: FOV= 320x320mm, voxel size= 1x1mm, slice thickness=3.5mm, gap= 0.125mm, TR/TE=1170ms, ip angel=90/110 (excitation/refocus) in 3 anatomical planes (15 2D stacks on average, range 8-31).Neonatal imaging was performed on 3 T Philips: Achieva-dStream: 3DMPRAGE T1 weighted images: FOV: 210x160x120mm, voxel size: 1x1x1mm, TR/TE 17ms/4.1ms, TI 1465ms.
3DSVR processing:
2DT2w HASTE stacks were reconstructed into 3D high-resolution volumes using NiftyMIC software (v0.8)[5]. Stacks with severe motion artefacts were visually identified and excluded. On average, 12 2D stacks were used (range 7-25). After reconstruction, NIFTI 3DSVR data were converted to DICOM and uploaded to a Philips Carestream workstation, identical to the local PACS workstation, for the assessment.
Analysis:
- Ventriculomegaly was assessed by a neuroradiology consultant and trainee with over 15 and 4 years of experience, respectively, using fetal 2D T2-weighted HASTE, fetal 3DSVR, and confirmed with 3D T1 neonatal scans.
- The visibility of 11 brain structures was evaluated using a 3-point scale: 0(not visible), 1(partially visible), and 2(clearly visible). The structures assessed included the posterior limb of the internal capsule (PLIC), pituitary stalk, optic nerves, olfactory bulbs, hippocampi, cortical grey-white matter differentiation, cortical folding, brainstem, deep grey-white matter contrast, and white matter lamination.
- Image quality was assessed by scoring the signal-to-noise ratio (SNR) on a 3-point scale: 0 (poor), 1 (medium), 2 (good). Motion artefacts were rated as 0 (no motion), 1 (mild motion), and 2 (severe motion). The Wilcoxon signed-rank test was used for statistical analysis. Of 20 subjects, eight had aqueduct stenosis identified on neonatal MRI. This was conclusively confirmed on 3/8 and 8/8 on 2D and 3DSVR, respectively, on fetal scans (Fig 1). There were no differences in image findings for the remaining ventriculomegaly diagnoses.
Fetal 3DSVR achieved higher visibility scores in 6 of the 11 rated brain structures (Fig 2). The statistically significant difference in mean score between 2D/3D was in PLIC (0.65vs1.85, p<0.001), Sylvian aqueduct (1.05vs1.9, p<0.001), olfactory bulbs (0.90vs1.70, p<0.01) and deep grey-white matter contrast (0.9vs1.9, p<0.01). Optic nerves and pituitary stalk showed no difference. The hippocampi, brainstem, and cortical folding were similar (Fig 3).
Fetal 3DSVR outperformed 2D in image quality assessment. SNR improved 35% of fetal scans (7/20) and motion artefacts were reduced in 25% of fetal scans (5/20)(Fig 4). On the antenatal ventriculomegaly cohort, fetal 3DSVR has demonstrated potential for enhancing the diagnosis of aqueduct stenosis by providing better visualisation and improved image quality in fetal brain MRI. Furthermore, the diagnosis was in agreement with neonatal ground-truth scans. This study offers evidence for integrating 3DSVR into clinical practice for antenatal assessment of ventriculomegaly and its use in clinical management. Future studies will expand on this work by validating 3DSVR in other fetal brain pathologies, further establishing its utility across various structural abnormalities.
Weaam HAMED (London, United Kingdom), Latha SRINIVASAN, Giles KENDALL, Leigh DYET, Donald PEEBLES, Anna L DAVID, Kelly Pegoretti BARUTEAU, Magdalena SOKOLSKA
16:30 - 16:40
#47933 - PG027 Reduced cerebral glucose metabolism in a mouse model of early-stage Alzheimer's disease detected by dynamic MR spectroscopy.
PG027 Reduced cerebral glucose metabolism in a mouse model of early-stage Alzheimer's disease detected by dynamic MR spectroscopy.
Alzheimer’s disease (AD) in humans and mouse models is characterized by decreased glucose uptake in the brain, according to FDG-PET studies[1]. In contrast, some FDG-PET investigations report no or increased FDG uptake both in human AD [2] and in mouse models [3,4]. This apparently hypermetabolic state occurs in the early-phase of AD, i.e. in mild cognitive impairment and young AD model mice [2,3,4,5,6]. To understand these discrepancies, additional methods dedicated to the in vivo assessment of glucose metabolism are needed. Deuterium MR Spectroscopy (DMS) and Imaging (DMI) using deuterated compounds have emerged as alternatives to FDG-PET[7,8,9]. These methods not only allow monitoring the uptake of glucose, but also absolute quantification and direct assessment of downstream metabolic conversions. This study aims to investigate glucose metabolism by dynamic DMS in the brain of an early-stage disease model, the APP/PS1 mouse at 6 months of age, which was shown to have increased FDG uptake [3,6].
We investigated 7 APPS/PS1 mice and 6 wild-type (WT) littermates (6 months old, weighting ~30 gram). Animals were anesthetized with isoflurane (in O2/air) and maintained at 37°C. Experiments were performed at 11.7T (BioSpec, Bruker BioSpin). Mice were positioned prone inside a volume 1H coil for background imaging and shimming. A custom-built 2H surface coil (12mm Ø; 76.8MHz) was positioned on the mouse head (Fig. 1). Baseline pulse-acquire 2H MR spectra were collected with TR=500ms and 300 averages (acquisition time 2:30 min). Next, the mice were infused in a tail vein with a 2 s bolus of 1.3g/kg deuterated glucose in saline. Subsequently 36 consecutive 2H spectra were collected for 90 mins. For postprocessing jMRUI 6.0 was used applying 2Hz line broadening and frequency alignment. The signals of deuterated water (HOD), glucose (Gluc), glutamine/glutamate (Glx), and lactate (Lac) were fitted with a Lorentzian line shape using AMARES [10]. Tissue concentrations of 2H metabolites were determined referenced to the natural abundance of HOD at baseline (13.7mM). For each mouse, metabolite concentrations were averaged from 10-60 min to calculate group averages for APP/PS1 and WT mice. The cerebral metabolic rate of glucose consumption (CMRgl) and TCA flux (Vtca) were determined from the dynamic 2H glucose, lactate and Glx data applying a one-compartment model (Cwave software [11]). Blood glucose levels and fractional enrichment as input functions were obtained from blood samples. In 2H MR spectra of WT and APP/PS1 mice, recorded immediately after application of deuterated glucose, a glucose 2H signal became visible next to the natural abundant 2H signal of HOD (Fig. 2). Shortly thereafter a combined signal for glutamine/glutamate and a signal for lactate appeared, which increased together with the HOD signal. After about 20 min the signals of glucose and lactate started to decrease, while that of HOD continued to increase (Fig. 3). The glutamine/glutamate signal reached a quasi-plateau at about 40 min (Fig. 3). The tissue concentrations of HOD, 2H glucose and lactate in the brain reached higher levels in the APP/PS1 mice than in the WT mice. Between 10-60 minutes after 2H glucose application the average glucose concentration was 6.39±0.43 mM for APP/PS1 and 5.28±0.46 mM for WT (p=0.0011) and the average HOD concentration was 19.38±0.47 mM for APP/PS1 and 17.80±0.35 mM for WT (p=0.0001). The average lactate concentration was 0.76±0.05 mM for APP/PS1 and 0.64±0.05 mM for WT (p=0.0012).
Kinetic analysis, fitting the 2H glucose, Glx and lactate data to a one-compartment model, yielded significantly lower CMRgl and Vtca for the APP/PS1 mice compared to WT mice (Fig.4). Finally, we found a significantly increased area of GFAP staining in the cortex. No Glut1 transporter area difference was found in the cortex and hippocampus. In the brain of the APP/PS1 AD mouse model at 6 months of age, glucose levels are increased compared to WT in agreement with Dynamic glucose enhanced MRI studies [12].
The reduced CMRgl explains the increased glucose levels. Thus, in contrast to previous FDG-PET studies we find decreased glucose metabolism in an early-stage Alzheimer disease model APP/PS1. Compared to FDG-PET, DMS or DMI has the advantage that downstream metabolism can also be assessed. The transiently increased lactate may be associated with astrogliosis, increased microglia and inflammation in agreement with an increased GFAP area in the cortex. The kinetic analysis also revealed a decreased Vtca which would agree with early mitochondrial defects in this mouse AD model [13]. In this study we demonstrate the potential of dynamic DMS to assess glucose in the brain of an Alzheimer mouse model, which may overcome the ambiquities of FDG-PET examinations. DMS and DMI have been shown to be feasible in the human brain [12] and thus may serve as clinical imaging biomarker in AD.
Andor VELTIEN, Sjaak VAN ASTEN (Nijmegen, The Netherlands), Maximilian WIESMANN, Tom SCHEENEN, Amanda KILIAAN, Arend HEERSCHAP
16:40 - 16:50
#47788 - PG028 Hypoxia-Targeted BOLD-MRI Reveals a Distinct Glioblastoma Tissue Response Extending Beyond the Contrast-Enhancing Tumor Border.
PG028 Hypoxia-Targeted BOLD-MRI Reveals a Distinct Glioblastoma Tissue Response Extending Beyond the Contrast-Enhancing Tumor Border.
Glioblastoma is the deadliest primary brain tumor and is characterized by abnormal neurovascular features and brain tissue infiltration extending beyond the contrast-enhancing(CE) tumor core. Current MRI techniques are suboptimal to accurately appreciate non-enhancing tumor tissue and the extent of glioblastoma infiltration. In the present prospective cross-sectional study, we investigated whether a new imaging contrast based on standardized transient hypoxic targeting during blood oxygenation level-dependent(BOLD)-MRI could be exploited to visualize glioblastoma tumor tissue and provide new insight on the peritumoral non-CE area.
Between April 2022 and November 2024 patients with radiological diagnosis of brain tumors were prospectively offered to undergo a BOLD-MRI during a standardized isocapnic double hypoxic protocol. %BOLD signal change, contrast-to-noise ratio(CNR), Rsquared and Lag were calculated voxel-wise in volumes of interest(VOI) using in-house written Matlab scripts after SPM preprocessing. Statistical analysis included comparison of calculated variables in tumor VOIs against contralateral flipped masks and of tumor VOI among each other and with healthy tissue. Color-coded overlay maps were produced for qualitative analysis. Twenty-six adult patients with newly diagnosed untreated glioblastoma were included (mean age 64.8 +/- 10 years, 20 male). Upon hypoxic stimulation, CE tissue displayed stronger negative %BOLD change and higher CNR with respect to contralateral flipped masks as well as higher Rsquared (all p<0.001) and longer lag (p=0.002). CE tissue was significantly different from GM for all parameters while from WM, edema and necrosis only for %BOLD signal change and CNR. Abnormal hypoxia-BOLD response extended to some non-CE FLAIR hyperintense peritumoral tissue in several subjects. In the present study we demonstrate that controlled hypoxic modulation during BOLD imaging robustly induces a tumor tissue contrast in patients with newly diagnosed glioblastoma. In particular, transient isocapnic hypoxia results in disproportionately stronger negative BOLD signal changes in CE tumor tissue, whereas healthy tissue, necrosis, and edema display a more modest response. Of note, while CE tumor’s response is to some extent heterogenous, evident signal change alterations extend in the FLAIR hyperintensity beyond CE tumor core in several of the included patients. According to the present understanding of the BOLD model, the potentiated negative signal response observed in glioblastoma CE tissue is to result from a higher, disproportionate, relative increase in local deoxyhemoglobin concentration upon hypoxic stimulation as compared to healthy one. This is likely to derive from a combination of higher CBV (stronger susceptibility effect during desaturation of higher blood volume), high resting CBF in absence of compensation to the stimulus (none to minimal vasodilation in isocapnic conditions, coupled to underlying inefficient autoregulation and impaired cerebrovascular reactivity i.e. CVR), inefficient modulation of oxygen extraction fraction(OEF) - and inability to adjust to changes in oxygen availability to restore metabolic equilibrium - and local aberrant anaerobic hypermetabolism. Our findings, complementing previous studies employing hypercapnic stimulus to explore tumoral and peritumoral CVR support the hypothesis that glioma-induced NVU, uncontrolled electrical activation, repetitive transient local hypoperfusion and hypoxia-driven aberrant vascular remodeling create a pathophysiological state of "dysfunctional hyperemia." In particular, we hypothesize that the physiological local transient functional hyperemic status whereby blood flow recruitment exceeds metabolic demand in healthy tissue is instead constitutively “active” at baseline state in glioblastoma due to hypoxia-induced neovascular features, abnormal electrical activation and synaptic integration of glioma cells into neural circuits as well as deranged local hypermetabolic changes, local shunting and dysfunctional oxygen processing. These neurovascular alterations would then contribute to result in a relatively higher local increase in deoxyhemoglobin during transient hypoxia in tumor tissue as compared with healthy one. Moreover, our preliminary findings suggest that, as abnormal BOLD response to hypoxia extends into specific non-CE peritumoral regions, this imaging contrast may provide critical insights into vascular changes preceding BBB breakdown, thus unmasking dysfunctional hyperemia and delineating tumor infiltration margins at an earlier stage than what appreciated with T1-CE MRI. Transient hypoxia induces a strong negative BOLD signal response with excellent model fit in glioblastoma tissue extending to some peritumoral area. As neurovascular alterations in peritumoral tissue precede blood brain barrier disruption, hypoxia-targeted BOLD-MRI may better depict glioblastoma infiltration beyond the CE tumor core.
Vittorio STUMPO (Zurich, Switzerland), Jacopo BELLOMO, Christian Hendrik Bas VAN NIFTRIK, Martina SEBÖK, Tristan SCHMIDLECHNER, Natalia CANTAVELLA FRANCH, Andrea BINK, Micheal WELLER, Zsolt KULCSAR, Luca REGLI, Jorn FIERSTRA
16:50 - 17:00
#47779 - PG029 Exploring intracellular environment in low-grade gliomas by diffusion-weighted MRS and APTw imaging at 3T.
PG029 Exploring intracellular environment in low-grade gliomas by diffusion-weighted MRS and APTw imaging at 3T.
Gliomas are aggressive primary brain tumors classified by IDH mutation and 1p/19q codeletion status: oligodendrogliomas (IDH-mutant, codeleted), astrocytomas (IDH-mutant, non-codeleted), and glioblastomas (IDH-wildtype)[1]. Diffusion MRI has been widely used to assess glioma microstructure and aggressiveness[2], but water diffusion reflects all tissue compartments, limiting specificity. Diffusion-weighted MR spectroscopy (dMRS) overcomes this by probing intracellular metabolite diffusion[3]. Reduced diffusion of neuronal tNAA reflects axonal degeneration[4], while increased diffusion of glial markers tCr and tCho suggests hypertrophy[5]. Amide Proton Transfer weighted (APTw) imaging, sensitive to mobile proteins, has also shown potential to differentiate IDH-mutant from wildtype gliomas[6]. In this study, we compared dMRS and APTw measures in gliomas versus healthy tissue, and between oligodendrogliomas and astrocytomas. We hypothesized greater diffusion of glial metabolites and higher APTw signal in astrocytomas, consistent with their distinct cellular morphology and greater aggressiveness.
Spectra were acquired with a single-voxel diffusion-weighted semi-LASER[7] sequence (TE/TR=120ms/3 cardiac cycles) at 3T (Siemens PRISMA). Two VOIs were located respectively within the tumor and in the contralateral tissue. Water suppression was performed with VAPOR and B0 shimming using FASTESTMAP[8]. Diffusion weighting was applied in three orthogonal directions with b ~ 3000 s/mm2 (16 averages per condition). Phase and frequency corrections on individual scans were performed before summation. Averaged spectra per diffusion condition were fitted with LCModel[9]. tCho, tCr and tNAA apparent diffusion coefficients (ADCs) were estimated from signal decays induced by the diffusion weighting, averaged on the 3 directions.
Data were acquired with B1 levels of 2 µT and 0.6 µT (saturation time: 2s, duty cycles: 90% and 50%)[10,11]. Frequency offsets varied from -6 to +6 ppm with increments of 0.5 ppm. Data processing was performed using Olea Sphere 3.0. Fluid-suppressed APTw maps were computed at amide offset frequency (3.5 ppm). Median Amide values were computed in the two VOIs.
Differences between healthy and glioma tissue were assessed using paired t-tests. Differences between gliomas subtypes were assessed with Welch’s t-tests. In vivo diffusion-weighted spectra are shown in Figure 1 together with FLAIR and amide maps in one patient. tCho and tCr ADCs were significantly higher in glioma than contralateral tissue (p=0.016 for both, Figure 2a). The ADC of tCr was significantly higher in astrocytomas (p= 0.045), but no other metabolites showed significant results in oligodendrogliomas (Figure 2b). Mean values of tCho and tCr ADCs were higher in astrocytomas than oligodendrogliomas, while the opposite trend was observed for tNAA ADC, however, these differences were far from statistical significance (Figure 2c).
Amide signal was significantly higher in high grade gliomas compared to lower grades. No differences in amide signal were observed between astrocytomas and oligodendrogliomas (Figure 3). Weak negative correlations were observed between amide signal and metabolite ADCs in oligodendrogliomas (Figure 4). Elevated tCr and tCho ADCs in gliomas vs contralateral tissue were consistent with two previous studies[12,13] performed in small cohorts of tumors and likely reflect the larger size of glioma cells compared to healthy cells, possibly combined with activated glia of the tumor microenvironment. The decreased tNAA ADC in astrocytomas may suggest a more severe axonal damage in this glioma subtype, compatible with their worse prognosis. Because metabolite intracellular diffusion should depend on both cellular morphology and intracellular properties such as viscosity and molecular crowding, we compared dMRS results with APTw measures in the same patients. tCr and tCho ADCs were increased despite the significant and expected increase in amide signal in tumor vs healthy tissue, and ADCs only weakly correlated with amide signal in oligodendrogliomas. These results suggest that protein accumulation in tumor may occur in different compartments (e.g. nuclei and mitochondria) than metabolite diffusion (cytosol and fibers). We explored tumor intracellular environment in a very homogeneous cohort of patients with low-grade gliomas by combining dMRS and APTw. Changes in dMRS metrics likely reflected the different morphology of glioma cells compared to healthy cells. More advanced acquisitions and modeling of dMRS data are needed to differentiate between oligodendrogliomas and astrocytomas based on their different microstructural properties.
Capucine CADIN (Paris), Stefano CASAGRANDA, Lucia NICHELLI, Bertrand MATHON, Marc SANSON, Stéphane LEHÉRICY, Małgorzata MARJAŃSKA, Francesca BRANZOLI
17:00 - 17:10
#47701 - PG030 Impact of Sweetened Milk on Teriflunomide Treatment in a Multiple Sclerosis Mouse Model: Disease Progression and Drug Reporting using Fluorine-19 MR.
PG030 Impact of Sweetened Milk on Teriflunomide Treatment in a Multiple Sclerosis Mouse Model: Disease Progression and Drug Reporting using Fluorine-19 MR.
Multiple sclerosis (MS) is a chronic autoimmune disease characterized by inflammation and neurodegeneration in the central nervous system (CNS) [1]. Experimental autoimmune encephalomyelitis (EAE) in SJL/J mice is a widely used animal model that replicates key features of relapsing-remitting MS (RRMS), enabling the evaluation of therapeutic interventions and drug delivery strategies. The pharmacokinetics, including bioavailability, of drugs of MS like teriflunomide (TF) may be influenced by the choice of delivery vehicle[2-4].
Recent advances in voluntary micropipette-guided drug administration (MDA) using palatable carriers, like sweetened milk (SM) and sucrose plus carboxymethylcellulose (SCMC), present promising alternatives to traditional dosing methods using forced gavage [5, 6]. As refinement, this approach improves animal welfare, aligning with 3R principles and minimizing stress-induced experimental variability.
In this study, we administered TF using voluntary MDA in EAE mice, using either SM or SCMC as delivery medium. We aimed to evaluate whether this delivery approach could ensure consistent intake while supporting hepatic accumulation and potential central nervous system (CNS) penetration of TF. We used ¹⁹F MR methods and high-performance liquid chromatography-mass spectrometry (HPLC-MS) to study TF MR properties and biodistribution [7].
EAE was induced in 24 female SJL/J mice via subcutaneous immunization with proteolipid protein peptide (PLP139-151) and assigned into vehicle and TF-treated groups (n = 6 per group). TF (30 mg/kg/day) was administered in either SM or SCMC, over 21 days. Body weight and clinical EAE score were recorded daily throughout the study.
At the end of the experiment, tissues (serum, brain, liver) were collected for ex vivo HPLC-MS and MR measurements. Ex vivo HPLC-MS tissue measurements and analysis for assessing the TF concentrations are done by Lipidomix GmbH. Excised liver and brain samples were fixed in 4% paraformaldehyde (PFA) and embedded in 2% agarose within 5 mL tubes for MR measurements using a Bruker 9.4 T MR system equipped with a ¹⁹F cryogenically cooled probe (¹⁹F CRP) and a 1H RF coil. Post-processing and statistical analyses were performed in MATLAB 2023b and R Studio. Mice treated with TF in SM (SM_TF group) exhibited no significant differences in disease incidence or overall severity compared to the SM groups (p > 0.05; Fig. 1a–b).
In contrast, administration of TF in SCMC markedly reduced disease severity, particularly between days 11 and 17 post-immunization (p < 0.05; Fig. 1a). Mice in the SCMC group began to show symptoms on day 9, and reached to peak on day 12 (Fig. 1 b). In the SCMC_TF group, we observed significantly reduced disease severity and delayed onset (p< 0.01; Fig. 1a–b).
Ex vivo HPLC-MS tissue analysis of the TF_SM group revealed, TF concentration levels in serum, liver, and brain consistent with previously reported findings (Table 1) [3]. The highest concentrations were observed in serum (78.24 ± 18.61 µg/g), followed by liver (64.11 ± 20.48 µg/g). TF levels in the brain were minimal (0.94 ± 0.52 µg/g).
¹⁹F MR spectroscopy of the TF_SM group (Fig. 2a) showed a TF signal peak at a chemical shift of -61 ppm in liver, in agreement with previous reports [3]. No detectable ¹⁹F MR signals were observed in the brain. ¹⁹F MR phantom experiments indicate that SM reduces the T2 of TF when compared to SCMC (Fig.3). This study demonstrates that TF treatment is markedly influenced by the choice of MDA medium. In sweetened milk, TF failed to reduce the disease severity of EAE, indicating that milk protein might interfere with the pharmacological properties of the drug. This outcome is consistent with the observed tissue-selective distribution pattern, in which TF preferentially accumulated in the liver. Both HPLC-MS and ¹⁹F MR spectroscopy confirmed low TF in the brain. One plausible explanation is that TF binds to milk proteins, thereby impairing its anti-inflammatory activity in lymphoid tissue. ¹⁹F MR relaxation experiments indicating a T2 shortening of TF in SM, suggest that milk protein might bind to TF also during in vivo applications, even though TF is mostly bound to serum proteins in the bloodstream.
These findings underscore the critical role of the administration vehicle in modulating drug biodistribution and efficacy, particularly in the context of neuroinflammatory diseases. Our findings demonstrate that the therapeutic efficacy of teriflunomide is strongly dependent on the choice of administration medium. While TF dissolved in sweetened milk failed to reduce EAE severity, TF administered in SCMC significantly reduced clinical symptoms. Further pharmacological investigations are needed to confirm these findings and to optimize TF delivery strategies for neuroinflammatory conditions.
Xiang HU (Berlin, Germany), Nandita SAHA, Yinhao CHEN, Jason M.MILLWARD, Michael ROTHE, Marc NAZARÉ, Friedemann PAUL, Thoralf NIENDORF, Sonia WAICZIES
|
Auditorium 900 |
|
"Friday 10 October"
Added to your list of favorites
Deleted from your list of favorites
B25
15:40 - 17:10
ET1-1 - Practical Statistics
ET1: Cycle of Research
15:40 - 17:10
AI as an Augmentation of Statistical Analysis in MRI: A Bridge Between Classical and Modern Approaches.
Alessia SARICA (PhD) (Keynote Speaker, Catanzaro, Italy)
15:40 - 17:10
Traditional and advanced statistics.
Ludovica GRIFFANTI (Keynote Speaker, United Kingdom)
|
Espace Vieux-Port |
|
"Friday 10 October"
Added to your list of favorites
Deleted from your list of favorites
C25
15:40 - 17:10
FT1 LT - Technological synergies in AI for MRI
Segmentation, reconstruction, and prediction
15:40 - 15:42
#47752 - PG187 Automatic segmentation and iron overload detection of myocardium from dark-blood T2* magnetic resonance images.
PG187 Automatic segmentation and iron overload detection of myocardium from dark-blood T2* magnetic resonance images.
Myocardial iron overload is commonly evaluated using T2* mapping from dark-blood cardiac magnetic resonance imaging [1-4]. Iron accumulation shortens T2* relaxation times, with a cut-off value of 20 ms at 1.5 T indicating pathological overload and an increased risk of cardiac dysfunction [5]. An accurate quantification requires manual segmentation of the myocardium, a time-consuming process prone to inter and intra-operator variability. To date, there’s a lack of a gold standard for both global and segmental myocardial T2* segmentation and several approaches based on both computer vision and deep learning have been applied [6-9]. Convolutional neural networks (CNNs) [10] can offer a promising solution for automating global and segmental T2* analysis. This study aims to develop and validate a CNN-based method for automatic segmentation of T2* dark-blood MR images, and to test its ability to detect myocardial iron-overload.
Three parallel short-axis slices of the left ventricle (basal, mid-ventricular and distal) were acquired with dark blood Multi-Echo Gradient Echo sequences at 1.5 T. Segmentations were manually delineated by an expert radiologist. Both antero-septal and infero-septal junctions of the inter-ventricular septum were identified to divide the myocardium into 16 equiangular segments, according to the American Heart Association (AHA) model [11]. Two segmentation tasks were considered: a global segmentation, distinguishing myocardium from background at the pixel level, and a segmental segmentation, assigning each myocardial pixel to one of 16 anatomical segments or to the background. The dataset provided is composed by 102 subjects, divided into 50 healthy subjects and 52 patients affected by pathologies such as hemochromatosis and thalassemia. The data was split into a training (N=77), and a test (N=25) sets. We selected two deep learning networks for our model to enhance robustness: 3D nnU-Net [12] and 3D U-Mamba_bot [13]. A 5-fold cross-validation training strategy was employed on the training set to optimize model performance. In each fold, the best-performing model was selected to generate segmentation predictions, that were then ensembled using STAPLE [14] algorithm (Fig.1 and Fig. 2). The performance of the model was assessed by using the Dice Similarity Coefficient (DSC) and the 95th percentile Hausdorff Distance (HD95). For clinical validation, T2* values were estimated in each segment using a mono-exponential model with a ROI-based approach [15]. Automated segmentations were compared against ground-truth contours on the test set to quantify agreement in T2* measurements. Using a 5-fold cross-validation, nnU-Net outperformed U-Mamba_bot in segmental segmentation, ranking best in 4 folds over 5. In global segmentation, nnU-Net achieved superior results in only 2 folds. Evaluation on the test set demonstrated a good agreement between manual and automated segmentations produced by the proposed algorithm for both tasks (Table 1). For segmentation overlap, the mean DSC was 0.819 ± 0.090 in the global approach and 0.738 ± 0.132 in the segmental approach. Regarding boundary accuracy, HD95 index was 2.000 ± 1.060 for global segmentation and 3.188 ± 2.573 for segmental segmentation. Average Pearson’s correlation of T2* values across the test set was 0.88±0.10. In the iron-overload classification task, the model achieved an accuracy of 0.88 in correctly identifying each of the 25 test subjects as either healthy or affected by iron overload, confirming the robustness and reliability of its predictive performance (Fig. 3). The proposed method can provide fully automated segmentation of dark-blood T2* images. Global myocardium segmentation achieved a better performance than segmental subdivision, since multi-class segmentation with a limited dataset dimension could be more challenging than binary global segmentation. In the segmental analysis, slightly lower performance was observed in the apical slice, likely due to increased noise and reduced image quality in that region. High segmentation accuracy was achieved in the mid-ventricular septum (segments 8 and 9), supporting the hypothesis that this area is less affected by artifacts [15]. From a clinical perspective, a key finding of our study is that average segmental T2* values estimated by our approach closely matched those obtained by expert radiologists as demonstrated by the achieved correlation. Misclassifications occurred for T2* values near the 20 ms cut-off, which lead to incorrect classification of iron overload. The proposed method provided good agreement with expert segmentations in both global and segmental myocardial segmentation. Furthermore, its performance in detecting myocardial iron overload is highly promising. These results lay the foundation for developing an advanced and fully automated pipeline for cardiac iron overload quantification, offering improved speed, reproducibility, and clinical applicability.
Ambra CHECCHETTO (Padova, Italy), Amalia LUPI, Giada BUSINARO, Simone PERRA, Alessandro GIUPPONI, Valentina VISANI, Alessia PEPE, Marco CASTELLARO
15:42 - 15:44
#45983 - PG188 A robust multi-network deep learning pipeline for automatic rectal cancer and mesorectum segmentation.
PG188 A robust multi-network deep learning pipeline for automatic rectal cancer and mesorectum segmentation.
Locally advanced rectal cancer (LARC) is a major cause of cancer-related morbidity and mortality [1] and accurate segmentation of the tumor and mesorectum on MRI is critical for treatment planning and therapy assessment [2,3]. Manual segmentation, however, is labor-intensive and prone to variability [4]. While deep learning methods have shown promise, most studies rely on single-scanner datasets or standardized protocols, limiting generalizability [5-9]. This study presents a robust deep learning pipeline for automatic segmentation of rectal tumors and mesorectum on T2-weighted MRI, trained and tested on a heterogeneous multi-scanner dataset. Data augmentation simulating typical MRI artifacts was applied, and final predictions were generated through an ensemble of segmentation models.
This study involved 141 patients (age: 61.6 ± 12.8 years; range: 27–87; M/F: 84/57) with LARC, imaged with axial T2-weighted MRI before neoadjuvant therapy. MRI scans were acquired across 17 different scanners (129 subjects at 1.5T and 12 subjects at 3T) from multiple institutions, introducing substantial variability in contrast, resolution, acquisition parameters and imaging protocols. This heterogeneity, along with anatomical variability of the rectal cancer and mesorectum, reflects real-world conditions and poses additional challenges for segmentation. Expert radiologists manually delineated both structures using 3D Slicer [10] (see Fig. 1 for examples of manual segmentations and variability in tumor shape).
The dataset was split into training (113 subjects) and testing (28 subjects) sets, maintaining scanner diversity by including at least one case from each scanner in the test set. Notably, 5 of the 17 MRI scanners contributed only one subject each to the entire dataset and these were all included in the test set to assess the model’s ability to generalize to previously unseen scanners.
Three different 3D network architectures were applied: nnU-Net [11], U-MambaBot [12] and Swin-UNETR [13]. Each model was trained using 5-fold cross-validation with identical splits. Training was conducted both with the standard nnU-Net data augmentation and with an extended scheme including TorchIO [14] and GIN-based [15] transforms to simulate typical MRI artifacts such as motion, ghosting, and bias field inhomogeneities.
Two ensemble strategies were evaluated. In the first, the five fold-based predictions of each architecture were combined, and the resulting three outputs were further aggregated using STAPLE [16]. In the second, for each fold, the model with the highest validation Dice score across architectures was selected, and its prediction on the test set was used for final aggregation (see Fig. 2 for a schematic overview of the training and testing pipeline of this latter approach).
Final segmentation performance was assessed on the test set using the Dice Similarity Coefficient (DSC) and the 95th percentile Hausdorff Distance (HD95), considering both rectal cancer and mesorectum. Four ensembling strategies were evaluated, differing in whether predictions were aggregated per architecture or per fold, and whether advanced MRI-specific data augmentation was applied. The best performance was achieved with the fold-wise ensembling strategy combined with extended data augmentation, resulting in a mean DSC of 0.706±0.132 and HD95 of 22.567±27.254 mm for the rectal cancer, and a mean DSC of 0.756±0.121 and HD95 of 9.677±6.119 mm for the mesorectum. Across all configurations, both data augmentation and ensembling consistently improved segmentation quality. All detailed results are reported in Table 3. Fig. 4 provides a qualitative example of model predictions compared to ground truth annotations, using the best-performing configuration. This study confirms the benefit of combining ensemble learning with MRI-specific data augmentation to enhance segmentation of rectal cancer and mesorectum on T2-weighted MRI. Augmentations simulating real-world artifacts improved both DSC and HD95, indicating greater robustness. Among the ensemble strategies, selecting the best-performing model per fold outperformed architecture-wise aggregation, likely due to improved exploitation of model diversity. Notably, Swin-UNETR was never selected in this setting, potentially as a result of the limited training data per fold, which may hinder transformer-based models known to require larger datasets [17-19]. This study highlights the effectiveness of integrating diverse 3D architectures, artifact-specific data augmentation and ensembling strategies for robust segmentation of rectal cancer and mesorectum on heterogeneous MRI. The use of a multi-scanner dataset improved generalizability, while STAPLE-based ensembling and advanced augmentations enhanced performance. Future efforts should focus on expanding data diversity, exploring additional imaging modalities and validating transformer-based models on larger cohorts to assess their full potential in clinical settings.
Simone PERRA (Padova, Italy), Filippo CRIMÌ, Valentina VISANI, Niccolò SION, Matteo PREZIOSO, Francesco CELOTTO, Claudio COCO, Giuditta CHILOIRO, Marco SCARPA, Emilio QUAIA, Simona DEIDDA, Gaya SPOLVERATO, Marco CASTELLARO
15:44 - 15:46
#47555 - PG189 Open-source multi-resolution graph cut algorithm for dual-echo water-fat separation.
PG189 Open-source multi-resolution graph cut algorithm for dual-echo water-fat separation.
Chemical shift-encoded water-fat separation is widely used for fat suppression and fat quantification in various anatomies. Dual-echo water-fat separation techniques allow efficient fat suppression, particularly in time-critical applications such as breath-hold acquisitions. Methods have been proposed that do not constrain the acquired echoes to in-phase and out-of-phase echo times allowing flexible echo timing [1-3]. Such methods usually exploit spatial neighborhood information to estimate a field or phasor map, similar to water-fat separation algorithms that require more than two echoes. However, the availability of open-source dual-echo water-fat separation techniques is still limited.
Previously, a multi-resolution graph cut algorithm was introduced for water-fat(-silicone) separation using more than two-echoes [4]. The algorithm demonstrated robust water-fat separation in the presence of B0 inhomogeneities with efficient processing times [4].
Building on this framework, this work presents an open-source dual-echo water-fat separation method based on a multi-resolution graph cut algorithm. The proposed method is designed to achieve robust water-fat separation in the presence of low SNR and large B0 inhomogeneities.
The proposed dual-echo water-fat separation algorithm applied a water-fat signal model neglecting T2* decay and assuming a common initial phase for the water and fat species [3]. The phase difference between the first and second echo represents the phasor map and is proportional to the underlying field map after appropriate correction. First, two possible phasor solutions were estimated corresponding to the real and the water-fat-swapped solution [3]. Second, the proposed multi-resolution graph cut algorithm was applied to estimate an unwrapped phasor map from two possible phasor solutions constraining the smoothness of the phasor map. Four different graph cuts with different spatial resolutions were performed to successively increase the spatial resolution of the phasor map (Fig.1). Third, water and fat images were estimated via matrix multiplication using the estimated phasor map [3].
The implementation is publicly available (https://github.com/BMRRgroup/2echo-WaterFat-hmrGC), and the examples presented in this study will be included in the repository. The algorithm requires as input the complex-valued signal at both echo times, a signal mask, and acquisition parameters such as echo times, field strength, and a predefined fat model.
Evaluation included a numerical simulation and in vivo datasets. The proposed multi-resolution approach was compared with a single-resolution graph cut used in the estimation of the unwrapped phasor map. A numerical Shepp–Logan phantom with varying fat fractions, linear field map and similar echo times compared to the in vivo scan was forward simulated at SNR of 10 and 100. In addition, the water–fat separation method was applied to 3D abdominal datasets with large field-of-view (FOV) coverage. The in vivo data were acquired at 3T (Ingenia Elition X, Philips Healthcare) using a free-breathing simultaneous water T1 and T2 mapping acquisition (2.5 mm isotropic resolution, FOV = 350 x 252.8 x 350 mm^3, TE = [1.0, 2.1] ms, TR = 3.4 ms), and water–fat separation was performed on the first subspace coefficient obtained after image reconstruction [5]. Water-fat separation employed a 9-peak in vivo fat model throughout the study [6,7]. Fig. 2 shows water and fat images and the field-map at SNR of 10 and 100, comparing the proposed multi-resolution and singe-resolution algorithms. Fig. 3 presents in vivo results for a large-FOV abdominal scan with a smoother field-map for the multi-resolution method, particularly at tissue boundaries. Fig. 4 shows additional volunteer data, demonstrating robust separation in the presence of large B0 inhomogeneities at the edge of the FOV. Results demonstrated the robust performance of the multi-resolution approach in the presence of low SNR for a numerical simulation and strong B0 inhomogeneities for a large-FOV abdominal scan. In comparison to the single-resolution approach, the multi-resolution graph cut algorithm is particularly well suited for the dual-echo water-fat separation problem due to incorrect phasor estimation in voxels with low SNR or at tissue boundaries. A dual-echo multi-resolution graph-cut algorithm was developed that is applicable at low SNR and in the presence of large B0 inhomogeneities. The algorithm is publicly available on Github.
Jonathan STELTER (Munich, Germany), Christof BOEHM, Dimitrios C. KARAMPINOS
15:46 - 15:48
#45708 - PG190 Comparison of Manual and Automated Segmentation of Human Articular Cartilage from 3D-MR Images: Influence of Field Strength and Knee Position.
PG190 Comparison of Manual and Automated Segmentation of Human Articular Cartilage from 3D-MR Images: Influence of Field Strength and Knee Position.
Accurate segmentation of articular cartilage is essential for assessing joint health and detecting early degenerative changes. While manual segmentation from high-resolution 3D-MRI offers precise anatomical detail, it is time-consuming and subject to inter-operator variability. Automated provide faster, standardized segmentation but may vary in performance depending on imaging conditions [1]. This study compares high-quality manual segmentation of human knee articular cartilage with automated segmentation by MRChondralHealth 4.1 (Siemens Healthineers), using 3D-MRI datasets (DESS- double echo steady state sequence) acquired at two magnetic field strengths (3T and 7T) and two knee positions (flexed and extended), to evaluate the influence of these variables on the segmentation accuracy and consistency.
Five volunteers were scanned at two whole body MRI scanners: 3T Prisma-Fit and 7T Investigational scanner (both Siemens Healthineers). Each subject was scanned twice: in fully extended knee position and in the knee flexion measured from long femoaral and tibial axis (22.5° in average) using 3D-DESS (double-echo steady state) sequence.
Each dataset was segmented with two different methods: fully automated CAN3D-based segmentation [2] (MR ChondralHealth 4.1, Siemens Healthineers) and manual segmentation performed by an experienced medical doctor resulting in four cartilage labels (“patella”, “femur”, “lateral and medial tibia”) and one concatenated label (“all”).
Three parameters were used to quantify the difference between manual and automated segmentation between field strength and knee positions, namely Dice coefficient (measures the overlap between two binary segmentation masks), Jaccard coefficient (quantifies the similarity between two sets by dividing the size of their intersection by the size of their union) and Hausdorff distance (measures the maximum distance from a point in one boundary set to the nearest point in the other boundary set). The differences of the means were compared using Student t-test, p-value lower than 0.05 was considered statistically significant. Comparison between manual and automated segmentation across magnetic field strengths revealed statistically significant differences, particularly favoring 3T over 7T imaging in terms of segmentation agreement. At 3T, the mean Dice coefficient across all cartilage regions was 0.84 ± 0.04, compared to 0.82 ± 0.04 at 7T (p = 0.02), with similar trends observed in the Jaccard index and Hausdorff distance. Notably, the Hausdorff distance showed a large discrepancy (100.95 ± 15.61 mm at 3T vs. 32.89 ± 46.35 mm at 7T, p < 0.001), suggesting greater boundary alignment issues at higher field strength. Region-wise, the femur and tibia (lateral) showed the most significant field strength-dependent differences, highlighting variability in segmentation robustness across anatomical regions. In contrast, comparisons between flexed and extended knee positions showed minimal statistically significant differences. Across all regions, Dice scores were similar (0.85 ± 0.03 for extended vs. 0.83 ± 0.09 for flexed, p = 0.12), and only the Hausdorff distance for the lateral tibia showed a significant difference (p < 0.001). This study highlights notable discrepancies between manual and automated segmentation of articular cartilage when comparing images acquired at different magnetic field strengths. The results demonstrated statistically significant differences in segmentation accuracy metrics (Dice, Jaccard, Hausdorff distance) between 3T and 7T datasets, particularly for automated segmentations generated by MRChondralHealth 4.1. These differences may be attributed to variations in image resolution, contrast, and noise characteristics inherent to different field strengths, which can influence the performance of automated algorithms.
In contrast, only minimal differences were observed between flexed and extended knee positions. This suggests that joint positioning has a relatively limited impact on the segmentation performance of both manual and automated methods, at least within the constraints of the current imaging protocols.
These findings underscore the importance of field strength consideration in both clinical and research applications of automated cartilage segmentation tools. While manual segmentation remains more consistent across imaging conditions, its time-intensive nature makes automation desirable—highlighting the need for field strength-adaptive or more generalized segmentation algorithms. Significant differences were found between manual and automated segmentation results when comparing MRI scans at 3T versus 7T, whereas knee position (flexed vs. extended) had minimal effect. These results emphasize the sensitivity of automated segmentation tools to variations in MRI field strength and the need for further optimization of such tools for reliable application across diverse imaging conditions.
Vladimir JURAS (Vienna, Austria), Veronika JANACOVA, Markus SCHREINER, Karin UNTERBERGER, Diana SITARCIKOVA, Pavol SZOMOLANYI, Esther RAITHEL, Gregor KOERZDOERFER, Siegfried TRATTNIG
15:48 - 15:50
#47882 - PG191 A Transformer Based Approach to Multi-modal Brain Tumor Segmentation with Arbitrary Missing Modalities.
PG191 A Transformer Based Approach to Multi-modal Brain Tumor Segmentation with Arbitrary Missing Modalities.
Follow-up of brain tumor treatment benefits from segmentation in Magnetic Resonance (MR) Images. However, manual segmentation of brain tumors is time consuming and labor-intensive. State-of-the-art automated brain tumor segmentation techniques typically rely on multiple magnetic resonance imaging (MRI) modalities, which, in practice, may not always be acquired in clinical settings. Moreover, contrast injections that are necessary to acquire gadolinium enhanced T1-weighted images, are contraindicated in certain patients (for example, patients with severe kidney failure) [1]. Therefore, developing more resilient deep learning segmentation models capable of handling cases with one or more missing modalities would be highly beneficial. Current leading methodologies addressing the missing modality challenge employ various strategies, such as training a separate model for each missing condition or leveraging fully available data during training while masking inputs to simulate missing modalities [2]. In this work, we introduce a novel transformer-based architecture [3] that integrates the available multi-modal features. The key advantage of our approach lies in its unified design, allowing it to process a variable number of inputs. The proposed method demonstrates competitive performance against state-of-the-art approaches in 2D whole tumor segmentation on publicly available Brain Tumor Segmentation Challenge (2021) data [4].
Three incremental methodologies were implemented and tested on the BraTS 2021 dataset [4], which consists of 4 co-registered MRI volumes of 1251 patients: native T1-weighted (T1), post-contrast T1-weighted (T1Gd), T2-weighted (T2), and fluid attenuated inversion recovery (T2-Flair); and respective ground truth (GT) segmentations validated by experienced radiologists: GD-enhancing tumor (ET), peritumoral edematous tissue (ED), and necrotic tumor core (NCR). All volumes were normalized and converted into 2D 128 x 128 axial slices. The GT labels were converted into 3 mutually inclusive regions that better represent the clinical application task: whole tumor (WT = ET + ED + NCR), tumor core (TC = ED + NCR) and enhanced tumor (ET, same as provided label) [5]. First, a baseline U-Net architecture, that requires all 4 modalities (4 channel inputs), was implemented as in [6] and adapted for 2D segmentation. As an intermediate step to address missing modalities, a modality dropout (ModDrop) [7] strategy was integrated into the baseline, by randomly dropping up to 3 modalities during training with a fixed probability. Our proposed approach (ModVit, Figure 1) integrates the previous methods with 2 major modifications: a single channel shared encoder that processes 1 available modality at a time as in [8] and a vision transformer [3] based bottleneck. The U-Net was trained with 4 modalities and tested in both full and missing modality conditions. ModDrop and ModVit were trained and tested with missing modality conditions, each modality was dropped with a fixed probability of 0.3 for each sample during training. The Dice loss was used as the minimization criterion and the Dice similarity coefficient as the main evaluation metric. In Table 1, Dice similarity scores are presented for the 3 tumor regions and 3 models tested in all possible missing modality configurations (i.e. fixed configuration repeated for every test sample). In the last test condition, a random configuration was attributed to each sample. In Figure 2, a qualitative comparison between the GT and model segmentations for U-Net and ModVit are presented for 2 axial slices, given a specific modality configuration (T1Gd and T2-Flair missing). Both quantitative and qualitative results show that ModVit is resilient to missing modality conditions, in contrast to U-Net. Despite achieving state-of-the-art Dice scores with 4 modalities, U-Net shows noticeably degraded performance in ET and WT segmentation with missing T1Gd and/or T2-Flair, which contain important information for the delineation of these regions by radiologists. This is also supported by the poor quality of segmentations in Figure 2. In ModDrop, randomly dropping modalities during training creates a regularization effect and constitutes a simple but effective method for making a network more robust to missing modalities, without changing its architecture. While the improvement of ModVit over ModDrop is incremental, it has the main advantage of accepting variable sized inputs and avoids mixing zeroed inputs with relevant data in the first layers, performing the feature integration only between available features in the bottleneck stage instead. In this work, we presented a flexible framework that addresses the missing modality problem, by integrating the variable sequence length processing capability of the transformer architecture, into an existing and validated methodology of automated brain tumor segmentation.
Marc GOLUB (Lisbon, Portugal), Rita G. NUNES, Carlos SANTIAGO, Jacinto C. NASCIMENTO
15:50 - 15:52
#47591 - PG192 Machine Learning-Based IDH Mutational Subgroup Classification in Gliomas Using Cerebral Blood Flow and BBB-ASL Exchange Times.
PG192 Machine Learning-Based IDH Mutational Subgroup Classification in Gliomas Using Cerebral Blood Flow and BBB-ASL Exchange Times.
Gliomas are the most common primary neoplasms in adults, and accurate histopathological grading and subtyping are essential for optimal treatment planning [1]. A key factor in glioma subtyping is isocitrate dehydrogenase (IDH) gene mutations [2]. Blood-brain barrier (BBB) breakdown is frequently observed in IDH-wildtype (IDH-wt) gliomas and brain metastases and is typically measured by assessing leakage of a gadolinium-based contrast agent on T1-weighted (T1w) MRI [3]. However, subtle changes in BBB integrity may be missed by conventional contrast-enhanced T1w MRI due to the higher molecular weight of the contrast agents [4]. The blood-brain barrier arterial spin labeling (BBB-ASL) technique provides a non-invasive alternative for detecting a broader range of BBB disruptions [5]. A previous study has demonstrated the feasibility of using BBB-ASL to identify IDH mutation status in gliomas based on cerebral blood flow (CBF) or exchange time (Tex) [6]. The current study improves this approach by jointly employing CBF and Tex using machine learning algorithms.
Twenty-five histopathologically proven gliomas (15 glioblastomas (IDH-wt), 10 astrocytomas (IDH mutant (IDH-mut)), M:F= 14:11, mean age= 53.6±14 years) were scanned on a clinical 3T MRI scanner (MAGNETOM Prisma, Siemens Healthineers, Erlangen, Germany) with a 32-channel head coil. A 3D TSE SPACE sequence (TR= 600 ms, TE= 12 ms, slice thickness= 0.8 mm, in-plane resolution= 0.4x0.4 mm2) was used to acquire pre- and post-contrast T1w MRI. FLAIR images were acquired using a 2D TSE sequence (TR= 9000 ms, TE= 92 ms, TI= 2400 ms, slice thickness= 3 mm, in-plane resolution= 0.7x0.7 mm2, spacing between slices= 3.6 mm). A combination of single-TE and multi-TE time-encoded pseudo-continuous ASL (pCASL) sequences was acquired with a 3D GRASE readout, implemented using gammaSTAR (details in Table 1) [7]. CBF and Tex were estimated using ExploreASL [8] with the two-compartment model implemented in FSL-FABBER [9].
Post-contrast T1w and FLAIR images underwent bias field correction (FSL FAST[10]) and skull stripping (FSL BET [11]). Whole tumor and necrosis regions were segmented using FLAIR and post-contrast T1w MRI in 3D Slicer [12]. Normal-appearing gray matter (NAGM) segmentations were performed on pre-contrast T1w MRI for the contralateral hemisphere using SPM12 [13]. Segmented masks were registered to CBF maps using SPM12.
Regional T2 values were obtained by fitting multi-TE control images from the Hadamard-4 PCASL to a mono-exponential decay using a non-linear optimizer in MATLAB, minimizing the least square error [14]:
S_{i}=S_{0}e^{-\frac{TE_{i}}{T2}}, i=[1:8] (1)
where S_{i} represents the i-th TE (TE_{i}) of the control images and S_{0} denotes the transverse magnetization signal. T2 and S_{0} were estimated simultaneously.
Patient-specific T2 values were then used to calculate corrected CBF (CBF_corr) and Tex (Tex_corr) maps. Finally, relative CBF_corr (rCBF_corr) and relative Tex_corr (rTex_corr) maps were generated by normalizing respective maps with the median T2-corrected values in NAGM.
To classify IDH-wt and IDH-mut tumors, eight histogram-based features—mean, median, standard deviation, 10th and 90th percentiles, skewness, kurtosis, and energy—were extracted from the rCBF_corr and rTex_corr maps. SHAP analysis was used to identify the top 10 among the total 16 features. Random forest, support vector machine (SVM), k-nearest neighbors (kNN), XG Boost, and naive Bayes classifiers were trained, and model performances were evaluated using three-fold cross-validation. Figure 1 shows rCBF_corr and rTex_corr maps for IDH-mut and IDH-wt patients. kNN performed best (accuracy: 0.833 ± 0.118, AUC: 0.859 ± 0.109), followed by random forest and XGBoost (accuracy: 0.759 ± 0.103 for both) with lower AUCs. SVM had lower accuracy (0.718 ± 0.066) but a strong AUC (0.827 ± 0.161) (Table 2). Overall, kNN outperformed the other models in terms of accuracy and AUC, making it the most effective model for this classification task. Figure 2 displays a) the SHAP summary plot and b) the kNN ROC curve. The machine learning analysis demonstrated that the kNN model outperformed all other classifiers across all performance metrics in distinguishing IDH-wt from IDH-mut gliomas. In addition to its superior classification performance, kNN was also among the fastest models, indicating both efficiency and robustness in this application. The complementary value of perfusion and exchange time metrics was evident, highlighting especially the exchange times as a promising imaging biomarker. SHAP analysis identified rTex_corr Energy and rCBF_corr Energy as the most important features. Although these results are encouraging, further validation is needed on larger datasets to confirm clinical applicability. Our findings support the potential of combining rCBF and rTex metrics with machine learning to improve non-invasive IDH mutation status prediction in gliomas.
Gülce TURHAN (Üsküdar, Turkey), Ayse Irem CETIN, Omer Yasin CUR, Beatriz E. PADRELA, Amnah MAHROO, Simon KONSTANDIN, Daniel Christopher HOINKISS, Nora-Josefin BREUTIGAM, Henk-Jan MUTSAERTS, Ayca ERSEN DANYELI, Koray OZDUMAN, Klaus EICKEL, Vera KEIL, Matthias GÜNTHER, Jan PETR, Alp DINCER, Esin OZTURK-ISIK
15:52 - 15:54
#47751 - PG193 Masked Auto-Encoders for Classification of Concussion using Imbalanced rs-fMRI Data.
PG193 Masked Auto-Encoders for Classification of Concussion using Imbalanced rs-fMRI Data.
Concussions are brain injuries diagnosed on subjective symptom reporting, and do not by definition result in gross structural brain changes. [1] However, rather than structural imaging, functional imaging such as resting state functional magnetic resonance imaging (rs-fMRI) has shown promise as a tool for concussion diagnosis and monitoring. Yet there are multiple rs-fMRI metrics, and which ones are most discriminatory between concussion patients and healthy controls remains unknown.
Deep learning models are becoming powerful tools for clinical decision making, but training these models assume class balance in the data. Unfortunately, medical data are often imbalanced for reasons such as privacy or rarity of conditions. This leads to biased models where higher accuracy is achieved by predicting the majority class because the samples are more likely to be from said majority class. This applies in concussion, where clinical cases are difficult to recruit and image while healthy control data (particularly from open sources) are abundant.
We proposed adapting a two-stage training process consisting of 1) pre-training a masked auto encoder (MAE) using a subset of the majority class (e.g., controls) and 2) training on the remaining, more balanced set of samples, for classification of concussion. We also analyzed the importance of features to identify important regions of interest (ROIs) for concussion classification.
Pediatric patients (n=28, aged 9-17yrs) who had been diagnosed with a concussion (within the past 4-weeks of injury) by a clinical partner experienced in pediatric concussion management were scanned using a GE Healthcare 3T MRI. Participants were excluded if they had more severe brain injury or prior neurological history. Healthy control data (n=500) of age matched subjects were obtained from the ABIDE-II[2] repository and were screened for data quality (i.e. signal-to-noise ratio, percentage of outlier scans, and data smoothness), resulting in 379 matched healthy controls.
Pre-processing was done using CONN 21a in MATLAB and functionalities of SPM12. The rs-fMRI data were functionally realigned and co-registered to the 3D T1-weighted anatomical data and warped into MNI space, slice-timing corrected, segmented and normalized, and spatially smoothed. Outlier detection was applied using SPM's Artifact Detection Tool and de-noising was applied using CONN's anatomical component-based noise correction. Temporal filtering was applied to remove frequencies outside of the 0.008-0.1Hz range. ROIs were then extracted using the Harvard-Oxford atlas, identifying 132 ROIs per subject. The data then had the following feature extraction methods applied to each ROI: mean, standard deviation, sample entropy[3], Lyapunov exponent[4], amplitude of low-frequency fluctuations (ALFF) and fractional ALFF(fALFF) [5] before being normalized.
The MAE closely follows the methodology of He et al. [6] while adjusting for use to address class imbalance. The MAE was pre-trained using 300 healthy controls to recover the original input from a subset of an input’s features generated by randomly masking (i.e. removing) portions of the input. The MAE consisted of an encoder of 5 transformer layers and decoder of 2 linear layers. After pre-training, the decoder was replaced with linear layers and re-trained on the remaining 79 healthy controls and 22 concussion patients for classification. Parameters for the linear layers were found empirically. Model performance and feature importance were assessed using a test set of 15 healthy controls and 6 concussion patients. Feature importance was identified through an ablation method where an input feature (ROI value) of a sample was set to -1 (an impossible value) and given to the model for classification. The difference between model output was then calculated. This was repeated for every input feature in the sample and all samples in the test set. The absolute value of the ablation analysis output across the test set was summed and ranked. The input features which change the model output the most were deemed important features. Figure 1 shows the output of the MAEs compared to the original input. Empirical classification results are detailed in Table 1 and show concussion classifiers trained with this method can have test accuracy up to 95.24% (sample entropy). ROIs deemed important for concussion classification include the vermis and cerebellar regions (Fig.2). The issue of imbalanced data is often addressed with synthetic data or transfer learning. Both methods have limitations such as overfitting from oversampling or the lack of previously trained models being available. This method does not require synthetic data or models trained on other data and results show that this may be an alternative to address the imbalanced nature of medical data. MAEs show promise for training a rs-fMRI based concussion classifier using imbalanced data, but more data is needed to solidify this finding.
Calvin ZHU (Hamilton, Canada), Bhanu SHARMA, Cameron NOWIKOW, Thomas DOYLE, Michael NOSEWORTHY
15:54 - 15:56
#45990 - PG194 Classification of Parkinson’s Disease Using Depthwise Separable 3D Convolutional Neural Network (DS-3DCNNs).
PG194 Classification of Parkinson’s Disease Using Depthwise Separable 3D Convolutional Neural Network (DS-3DCNNs).
Parkinson's Disease (PD) is a neurodegenerative disorder characterized by the degeneration of dopaminergic neurons in the substantia nigra, leading to motor and cognitive impairments [1]. Magnetic Resonance Imaging (MRI) plays a vital role in PD diagnosis by providing detailed soft tissue contrast and three-dimensional structural information [2]. Traditional PD diagnosis often relies on manual interpretation of MRI scans, which is subjective and time-consuming. Deep learning, particularly Convolutional Neural Networks (CNNs), offers an automated and systematic method for feature extraction from MRI data [3].
3D CNNs offer significant advantages over 2D CNNs in processing volumetric MRI data, capturing both spatial dependencies and volumetric information. However, 3D CNNs also present challenges, such as high computational costs and memory demands, leading to extended training times and resource inefficiencies. Additionally, the large number of parameters increases the risk of overfitting, particularly with smaller datasets. To address these, we used depthwise separable convolutions (DS-CNNs), reducing parameters by 10-20%, improving efficiency and mitigating overfitting without sacrificing performance [4]. This makes 3D CNNs more feasible for high-dimensional medical imaging tasks, such as PD detection.
While CNNs have shown success in detecting PD at later stages, research focusing on early-stage or prodromal PD detection remains limited. This study aims to address this gap by developing a deep learning-based system that incorporates the prodromal phase into the diagnostic pipeline. By leveraging DS-3DCNNs, this study seeks to enhance the diagnostic precision of early-stage PD detection, ultimately enabling more effective early interventions.
This study focuses on detecting Parkinson’s Disease (PD) and classifying MRI scans into three categories: healthy control, prodromal, and Parkinson’s Disease, using depthwise separable 3D convolutional neural networks (DS-3DCNNs). The methodology consists of four stages: MRI scan acquisition from the PPMI database, data preprocessing and registration, DS-3DCNN architecture, and performance evaluation. The DS-3DCNN architecture efficiently extracts spatial and volumetric features from high-dimensional MRI data, reducing computational cost without compromising performance. Each convolutional layer is followed by a ReLU activation and max-pooling to minimize overfitting. The final output layer uses softmax activation for classification into PD, healthy control, or prodromal stages. The model, with 3,094,115 parameters, was trained on 426 subjects from the PPMI dataset [5]. Image registration was performed using ANTsPy for spatial consistency. The method is detailed in Figure 1, and the architecture is shown in Figure 2. The performance of the proposed 3D Convolutional Neural Network (3D CNN) model was evaluated using 5-fold cross-validation. The model demonstrated strong classification accuracy across three categories: Parkinson's Disease (PD), Healthy Control (NC), and Prodromal. The average accuracy was 91.0 ± 2.2%, with Split 3 achieving the highest accuracy of 93.45%. Split 3 also achieved a precision of 0.9420, recall of 0.9298, and an F1-score of 0.9284, with an ROC-AUC of 0.94, indicating strong discriminatory ability, as shown in Table 1. These results demonstrate consistent performance across all splits with low standard deviations.
To further assess model interpretability, Grad-CAM was applied to visualize the regions of the MRI scans that most influenced classification decisions. For the PD class, activations were observed in key areas such as the basal ganglia and brainstem, which align with known PD pathology, as shown in Figure 3. In the Prodromal class, the model highlighted regions indicative of early neurodegenerative changes, supporting the model’s alignment with established neuroanatomical patterns of PD progression. The results demonstrate that the DS-3DCNN model effectively classifies Parkinson’s Disease, healthy controls, and prodromal cases, achieving an accuracy of 91.0 ± 2.2% and precision of 0.922 ± 0.015. The model's ability to identify key regions associated with PD pathology, such as the basal ganglia and brainstem, enhances its clinical relevance. Despite challenges in early-stage detection, the model's strong performance underscores its potential for early intervention, supporting the integration of DS-3DCNNs into clinical diagnostic pipelines. This study demonstrates the effectiveness of DS-3DCNNs in detecting Parkinson's Disease, particularly in the prodromal stage, using MRI scans. The model’s high accuracy and ability to identify biologically relevant regions offer promising implications for early diagnosis and intervention. Further validation and integration of this model could significantly improve early-stage PD detection and patient outcomes in clinical settings.
Muhammad ZUBAIR (Chieti, Italy), Matteo FERRANTE, Cosimo DEL GRATTA, Nicola TOSCHI
15:56 - 15:58
#47391 - PG195 Differentiation of parkinsonian syndromes based on multimodal MRI and 3D convolutional neural network.
PG195 Differentiation of parkinsonian syndromes based on multimodal MRI and 3D convolutional neural network.
Multiple system atrophy (MSA), a rare atypical parkinsonian syndrome, can prove challenging to differentiate from Parkinson’s disease (PD), especially at an early stage [1]. Combining MRI data and machine learning techniques has shown great potential in aiding differential diagnosis [2]. As a powerful tool for image analysis, convolutional neural networks (CNNs) enable the analysis of multidimensional images, such as MRI, offering an automatic and user-independent tool [3]. This study advances using a 3D CNN to distinguish patients with MSA from those with PD relying on multimodal, multicentric MRI data. CNN predictions were investigated by highlighting the most important regions and examining the characteristics of misclassified patients.
The patient cohort included patients with PD and patients with MSA presenting the cerebellar (MSA-C), parkinsonian (MSA-P), and mixed (MSA-PC) variants. We gathered MRI data from three sites of the French MSA reference center (Bordeaux, Paris, Toulouse) acquired with 3T MR scanners. For our multimodal pipeline, we considered the T1-weighted sequence, to compute gray matter density (GD) maps, and diffusion tensor imaging data, to compute mean diffusivity (MD) maps. All images were normalized in the MNI space with a 2×2×2 mm3 resolution. While GD maps give insights into macrostructural changes, e.g. atrophy, MD maps inform about the microstructural integrity of cerebral tissues. GD and MD maps were used as input individually or combined into a three-dimensional CNN architecture [4] to differentiate between PD and MSA or its variants. We trained the CNN with a 50-time repeated five-fold cross-validation on 80% of the dataset, the remaining used as a hold-out set for testing, considering three different data splits. We assessed performances with common evaluation metrics (sensitivity, accuracy, sensitivity) on the hold-out sets. To improve model interpretability, we (i) employed a visualization technique [5] to determine the most relevant regions for prediction, and (ii) examined the clinical and imaging characteristics of misclassified patients. The patient population included 64 patients with PD and 92 patients with MSA, comprising 9 with MSA-PC, 33 with MSA-C, and 50 with MSA-P [2,6–9]. The CNN yielded the best accuracies using MD for the PD vs MSA-C/PC task (0.84±0.08), GD for the PD vs MSA-P task, and both GD-MD maps for the PD vs MSA (all variants) task (0.88±0.03). We observed a gap between sensitivity (performance on MSA patients) and specificity (performance on PD patients), the latter superior to the former (0.71-0.84 vs 0.75-0.99). Milder and fewer image alterations emerged for misclassified patients, who presented overall younger age and shorter disease duration. The visualization technique highlighted as relevant for CNN prediction, regions involved in the MSA pathophysiology, i.e. the cerebellum and putamen. This study successfully differentiated MSA from PD patients using an MRI-based pipeline via a 3D CNN. While the monomodal approach gave insight into the informative content of a single MRI map (GD or MD), the bimodal approach (GD-MD) improved overall performance. This could be attributed to the complementary information from the two MRI maps, reinforcing the advantage of multimodality [10]. The most challenging task was PD vs MSA-P, whereas increased sensitivity was found for the others. A key requirement for AI in medical care is the need for transparency and elements of interpretability [11]. In this regard, we investigated CNN performance by exploring convolutional layer activations resulting in a visual interpretation. Interestingly, we found regions of interest in the differentiation of MSA from PD which were the most activated, thus supporting the CNN predictions. Albeit blind to clinical characteristics, the CNN seemed more prone to misclassify younger patients with a shorter disease duration. This warns about the necessity of developing automatic tools to distinguish early-stage data. Moreover, the analysis of misclassified patients from an image point of view showed that they were characterized by less severe and fewer alterations, hence the difficulty for the CNN to capture these subtler anomalies. Although promising, these findings come with some limitations. First, due to the rarity of the MSA, our study is burdened by data paucity. However, gathering multicentric data and keeping MRI parameters as homogenous as possible, represented a great effort and favored a fair disease heterogeneity. Second, disease confirmation can only be obtained post-mortem and is seldom available. Future work includes validating our approach using external data and extending to other parkinsonian syndromes. This study marks the beginning of a comprehensive exploration of the potential of MRI combined with CNNs for differentiating parkinsonian syndromes. Our objective is to develop an image-based automated aid-to-diagnosis tool.
Giulia Maria MATTIA (Toulouse), Lydia CHOUGAR, Alexandra FOUBERT-SAMIER, Wassilios G. MEISSNER, Margherita FABBRI, Anne PAVY-LE TRAON, Olivier RASCOL, David GRABLI, Bertrand DEGOS, Nadya PYATIGORSKAYA, Alice FAUCHER, Marie VIDAILHET, Jean-Christophe CORVOL, Stéphane LEHÉRICY, Patrice PÉRAN
15:58 - 16:00
#47885 - PG196 Exploring Kolmogorov–Arnold Networks for 3D T1-weighted MRI-Based Brain Age Prediction.
PG196 Exploring Kolmogorov–Arnold Networks for 3D T1-weighted MRI-Based Brain Age Prediction.
Brain age prediction from T1-weighted MRI is a key biomarker of neurological health, sensitive to neurodegeneration [1-3] and brain development [4]. Convolutional Neural Networks (CNNs) have been widely adopted for this task, given their ability to extract spatial features from MRI scans [5–7]. However, Kolmogorov–Arnold Networks (KANs), inspired by the Kolmogorov–Arnold representation theorem, have recently demonstrated competitive or superior performance in various image-related tasks [8, 9]. KANs approximate complex functions more efficiently than conventional neural networks and have shown promise in classification, segmentation, and generative tasks. Here, we present the first application of KANs for 3D brain age prediction and compare their performance to standard CNNs.
We used T1-weighted MRI scans from three publicly available datasets: the Human Connectome Project [10], the Nathan Kline Institute - Rockland Sample [11], and the Cambridge Centre for Aging and Neuroscience [12]. The combined cohort included 2,129 participants (878 males and 1,250 females), ranging in age from 18 to 100 years. All images were linearly co-registered to MNI152 2009c standard space to ensure spatial alignment and a uniform input shape (193×229×193). To enhance model generalizability, we applied data augmentation, consisting of rotation (±40°) and translation (±10 pixels) [13]. Data were randomly split into training (64%), validation (16%), and test (20%) sets, stratified by age and sex. For cross-validation experiments, the training and validation sets were redefined in each fold. Mann-Whitney U tests confirmed no statistical differences in age or sex between training and test sets (p = 0.901) nor between training and validation sets across cross-validation folds (lowest p = 0.840).
We evaluated: a baseline CNN [14], a convolutional KAN with a linear KAN output layer (KAN), and a hybrid CNN with a final linear KAN layer (CNN + KAN-Lin). Both CNN and hybrid models used 3×3×3 kernels and strides of 1 and 2 in the first convolutional layer. For memory efficiency, the KAN model was only tested at stride 2. All models were trained using MSE loss and the Adam optimizer (learning rate = 0.0001) for 1000 epochs, with validation every 50 epochs. The best models, those with the lowest validation loss, were assessed on the test set using Mean Absolute Error (MAE) and Pearson Correlation Coefficient (r). In cross-validation, performed only for stride-2 models, final predictions were obtained via median ensembling of the best-performing models from each fold. As shown in Table 1, KAN model with stride 2 reduced MAE by 15.16% over the baseline CNN, while the hybrid CNN + KAN-Lin model with data augmentation (DA) improved MAE by 11.72%, offering the best trade-off between accuracy and computational load. Moreover, DA consistently improved generalization across models, improving the performance on unseen test data. Due to memory constraints and hybrid’s model efficiency, stride-1 evaluation excluded the KAN model. At stride 1, the hybrid model outperformed the CNN baseline by 5.77% with DA. However, the performance gain was smaller than with stride-2, likely because high-resolution input allowed CNN layer to extract finer features, reducing the added value of the KAN layer.
Despite improvements, all models exhibited age bias (Figure 1): younger subjects’ ages were overestimated and older subjects’ underestimated. However, the hybrid model showed a smoother Predicted Age Difference (PAD) curve and improved accuracy for middle-aged individuals, though biases persisted at age extremes. These findings suggest that KANs offer a promising alternative to CNNs for brain age prediction from 3D MRI. In particular, the hybrid CNN + KAN-Lin model emerged as the most efficient and accurate configuration, effectively combining CNNs’ spatial feature extraction with KANs’ expressive function approximation. Moreover, DA played a crucial role in enhancing model robustness. However, persistent age bias highlights the need for additional strategies such as post processing debiasing methods or regularization. The high computational demand of KANs remains a limitation for high-resolution inputs. This study demonstrates, for the first time, the potential of KANs for brain age prediction from 3D MRI. A hybrid CNN + KAN-Lin architecture achieved the best compromise between predictive accuracy and computational feasibility, showing consistent generalization with data augmentation. While both models exhibited age-related bias, the hybrid approach produced smoother PAD distributions and better accuracy for mid-life age ranges. This synergy of CNNs and KANs opens a new direction for efficient, interpretable, and generalizable neuroimaging models. Future work will focus on mitigating age bias and scaling pure KANs for full-resolution inputs.
Alessandro GIUPPONI (Padova, Italy), Davide DE CRESCENZO, Marco PINAMONTI, Valentina VISANI, Manuela MORETTO, Alessandra BERTOLDO, Mattia VERONESE, Marco CASTELLARO
16:00 - 16:02
#46042 - PG197 Gestational Age Prediction with Transfer Learning and Pre-Trained CNNs Architectures using Fetal MRI Data.
PG197 Gestational Age Prediction with Transfer Learning and Pre-Trained CNNs Architectures using Fetal MRI Data.
Gestational age prediction is crucial in prenatal care, aiding in fetal development assessment and health risk evaluation. Traditional methods like ultrasound (US) are commonly used but can be inaccurate, especially when imaging quality is compromised by maternal obesity, fetal positioning, or low amniotic fluid levels [1]. US also has limitations in later pregnancy stages, with errors of up to 2–4 weeks.
Recent advancements in medical imaging, particularly Fetal MRI, improve gestational age estimation. Fetal MRI provides superior resolution, enabling detailed visualization of fetal brain anatomy and myelination, essential for accurate predictions [2]. However, challenges such as rapid developmental changes, suboptimal signal-to-noise ratios, geometric distortions, and fetal motion affect image quality [3]. Additionally, inconsistencies in MRI protocols and operator expertise complicate interpretation.
Deep learning, particularly Convolutional Neural Networks (CNNs), is a powerful tool for analyzing complex medical images. CNNs excel at extracting detailed features from large datasets, making them ideal for tasks like gestational age prediction from fetal MRI. Transfer learning, which fine-tunes pre-trained models on smaller datasets, addresses data limitations in medical imaging [4]. This study explores transfer learning with pre-trained CNN architectures to improve gestational age prediction, aiming to enhance accuracy and provide more reliable estimates.
This study used transfer learning with the VGG16 architecture to predict gestational age from fetal MRI scans. The dataset, sourced from ITAB, includes 261 studies with T2-weighted MRI sequences across axial, coronal, and sagittal planes. Preprocessing involved converting DICOM images to JPG, resizing them to 224x224 pixels, and converting grayscale to RGB. Data were segmented by anatomical plane, with key slices (1, 3, and 5) selected for optimal fetal brain representation.
The VGG16 model was fine-tuned using transfer learning. Initially pre-trained on ImageNet, the model was adapted for gestational age prediction by replacing classification layers with regression output layers, as shown in Figure 1. Experiments included single-planar and multi-planar approaches. In the single-planar phase, models were trained on individual anatomical planes, while in the multi-planar phase, data from all three planes were combined. Attention mechanisms were incorporated to focus on the most relevant areas of the MRI scans. The transfer learning process adapts a pre-trained architecture to our task, leveraging features from ImageNet. The model is trained end-to-end to predict gestational age from fetal MRI scans [5]. We developed a pre-trained VGG16 network using transfer learning to improve gestational age prediction from fetal MRI scans. The dataset included MRI scans from axial, coronal, and sagittal planes, with slices of 1, 3, and 5 selected for optimal representation of key brain features.
The VGG16 model was fine-tuned, and an attention mechanism was incorporated to enhance performance. Models were evaluated using R² scores and Mean Absolute Error (MAE) in days. As shown in Table 1, multi-planar configurations outperformed single-planar setups. Specifically, VGG16 demonstrated the best performance with 5-slice multi-planar input, achieving an R² score of 0.9461 and an MAE of 5.35 days, indicating strong predictive power. The attention mechanism further enhanced performance, reaching an R² score of 0.9581 and an MAE of 4.51 days, as shown in Table 1. These results confirm that the VGG16 architecture, especially with the attention-guided approach, is highly effective in predicting gestational age from fetal MRI scans, leveraging complementary anatomical information for more accurate predictions. The results demonstrate the effectiveness of the VGG16 architecture, particularly with multi-planar data and attention mechanisms, in accurately predicting gestational age from fetal MRI scans. The model's performance significantly improved with the incorporation of multiple anatomical planes, showcasing the value of leveraging complementary features for accurate predictions. The attention-guided approach further enhanced performance, achieving the highest results of R² = 0.9581 and MAE = 4.51 days with the multi-planar attention mechanism, focusing on critical brain regions and increasing robustness. The VGG16 model, enhanced with transfer learning and attention mechanisms, provides highly accurate gestational age predictions from fetal MRI scans. Multi-planar data integration significantly improves model performance, highlighting the importance of leveraging detailed anatomical information for precise predictions. These findings support the potential of deep learning techniques in advancing prenatal diagnostics and providing reliable clinical tools for gestational age estimation.
Muhammad ZUBAIR (Chieti, Italy), Massimo CAULO, Cosimo DEL GRATTA
16:02 - 16:04
#47624 - PG198 Predicting a Personalized Reference Model of Lung Dynamics from 3D MRI Using Deep Learning.
PG198 Predicting a Personalized Reference Model of Lung Dynamics from 3D MRI Using Deep Learning.
Clinical diagnosis often involves comparing patient data to reference models. In the case of lung dynamics, a generic model would offer limited clinical value given the high inter-subject variability. To address this, we propose generating a personalized dynamic reference that allows for more accurate and meaningful assessments. The present work aims to predict subject-specific volumetric lung deformations from binary masks. The resulting framework produces a personalized 4D reference to enable comparison with the patient's actual breathing dynamics.
The European project V|LF-Spiro3D aims to develop and validate free-breathing 3D MR spirometry, a technique originally introduced by Boucneau et al [1] to dynamically capture lung motion, characterize function, and map ventilation throughout the respiratory cycle. Unlike traditional spirometry, which uses forced expiration and yields only global measurements, 3D MR spirometry allows regional and local analysis under natural, free-breathing conditions.
This work leverages 4D MRI scans acquired from 50 healthy volunteers under the V|LF-Spiro3D clinical protocols, including both supine and prone positions across 32 respiratory phases. The lungs are segmented using the pipeline developed by Barrau et al. [2]. The complete dataset comprises 224 images and is split 60%, 20%, 20% into training, validation, and test sets, with no subject overlap between sets.
To capture both anatomical shape and spatio-temporal dependencies, we extended the deep learning architecture proposed by Vaurs et al. [3], which combines a 3D CNN encoder–decoder with an LSTM. The model (fig.1) includes skip connections from the first phase (end-expiration), and from the current phase (last frame of each sub-sequence).
To enhance the original architecture, we refined it by incorporating additional components. First a skip connection from the sixteenth respiratory phase was added, typically corresponding to peak inspiration. This provides a direct structural reference for target lung expansion. In a separate approach, tidal volume (TV), defined as the volume difference between the first phase (end-expiration) and the phase of maximum expansion, was introduced into the latent space. This scalar aimed to replicate the information conveyed by the skip 15 image, while maintaining the goal of predicting the full respiratory cycle from a single input phase. Different configurations were evaluated to assess the contribution of each component, including A (skip 0+skip current), B (skip 0+skip 15), C (skip 0+skip current+TV), and D (skip 0+TV). Models were trained by optimizing the binary cross-entropy (BCE) loss which measures voxel-wise classification accuracy. In addition to BCE, performance was evaluated using several metrics: relative volume error, to quantify differences in total lung volume between prediction and ground truth; Dice score to assess volumetric overlap; 95th percentile Hausdorff Distance (HD95) and Average Symmetric Surface Distance (ASSD) to assess surface accuracy.
Among the four configurations, B performed best across all metrics (figs.2&3), followed by D with slightly lower scores. Configurations A and C performed worst, though C showed marginally better results, likely due to additional physiological information from TV. As expected, errors increased at phases farther from the skip phases, however, skip connections from the current phase did not improve performance. Fig.4 illustrates 3D predictions using B on a test sample for visual assessment. Anchoring the network at both respiratory extremes significantly improved prediction accuracy. Incorporating TV enhanced personalization by capturing individual variability without requiring additional imaging, though slightly less accurate than with the peak-inspiration image, results remained satisfactory.
In future work we will apply the model for patients suffering from chronic obstructive pulmonary disease (COPD) and asthma. The predicted dynamic will be compared to the patients’ actual respiratory motion to detect deviations that are potentially linked to disease-related abnormalities.
Additional strategies will also be explored to improve temporal generalization, such as scheduled sampling, which gradually replaces ground-truth inputs with model predictions during the training phase. Furthermore, demographic data (height, weight, age, gender) will be incorporated to enhance the model's ability to capture subject specific characteristics.
Finally, we will build upon the current model to develop an enhanced version that can operate on grayscale images, enabling a more detailed structural representation beyond binary masks. The proposed framework achieves accurate prediction of full 4D lung shape dynamics from a single image at end expiration, with low volumetric and surface errors across healthy subjects. These results highlight the models’ strong potential as a patient-specific reference for identifying abnormal respiratory patterns.
Georges ABOU MRAD, Damien VAURS, Xavier MAITRE, Dima RODRIGUEZ (Paris-Saclay)
16:04 - 16:06
#47554 - PG199 Disentangled Forward-Distortion Network for Distortion Correction in EPI.
PG199 Disentangled Forward-Distortion Network for Distortion Correction in EPI.
Susceptibility-induced distortions in echo planar imaging (EPI) can severely deteriorate image quality, affecting the performance of diffusion-weighted imaging (DWI) and functional MRI where EPI is commonly used [1,2]. Classical methods use reverse phase-encoded (PE) EPI images to estimate an anatomically correct image together with an underlying off-resonance field [3]. However, these methods suffer from long computation times due to their iterative nature, making them impractical in clinical settings.
Recent advancements have focused on deep learning methods to speed up EPI distortion correction [4-5]. We previously introduced an unsupervised forward-distortion network (FD-Net) that rapidly estimates a corrected image and a displacement field using a 2D U-Net [6]. The predicted image is then forward distorted in reversed-PE directions using the estimated field. Physics-driven data consistency is enforced between the forward-distorted images and the input images for unsupervised learning. Using a single U-Net encoder-decoder architecture for simultaneously estimating a corrected image and a field map can cause anatomical details to leak into the field map. In this work, we propose Disentangled Forward-Distortion Network (DFD-Net), featuring disentanglement units in the encoder that split feature maps into separate streams that feed two separate decoders for image and field prediction. DFD-Net significantly improves distortion-correction fidelity and field map accuracy compared to FD-Net, while delivering over 200-fold computational speedup over TOPUP.
Proposed DFD-Net: To prevent anatomical image details from leaking into the displacement field, the proposed DFD-Net features a disentanglement unit that splits selected feature maps into separate streams that feed two separate decoders (see Figure 1). In the disentanglement unit shown in Figure 1(c), each selected feature map is processed via reduced singular value decomposition (SVD). Assuming larger singular values contain the majority of the high-resolution anatomical details, the largest m singular values are assigned to the image prediction stream, and the remaining ones are assigned to the field prediction stream. Here, the tunable parameter m controls the amount of activation information channeled into the image versus the field streams. By providing each decoder with disentangled skip-connections and bottleneck features, the anatomical content is forced to remain in the predicted image and not contaminate the displacement field.
Learning Procedures: We used randomly selected unprocessed DWI data from the Human Connectome Project’s 1200 Subjects Data Release [7]. A total of 24 subjects were selected, split as (12,4,8) subjects for (training, validation, testing). All b0 volumes were utilized, consisting of 111 slices, 168x144 image matrix, and 6 repetitions. We compared DFD-Net with (1) FD-Net and (2) a supervised baseline that is trained using TOPUP results. All models employed the same set of hyperparameters for fair comparison. Quantitative evaluations were performed on the corrected images and field maps using PSNR and SSIM metrics, taking TOPUP corrected images and field maps as reference. For each volume, distortion correction took ~11 seconds for DFD-Net and ~4 seconds for FD-Net, compared to ~37 minutes for TOPUP. Therefore, DFD-Net provides over 200-fold speedup compared to TOPUP.
First, we varied the number of singular values assigned to the image prediction stream at different resolution levels within the encoder. As shown in Figure 2, the accuracy of DFD-Net is more sensitive to the choice of singular values at the higher resolution levels.
The quantitative assessments in Figure 3 show that DFD-Net provides 2.13 dB PSNR and 5.71% SSIM improvement in field quality and 0.44 dB PSNR and 1.46% SSIM improvement in image quality over FD-Net. In addition, DFD-Net outperforms the supervised baseline with 2.06 PSNR and 21.99% SSIM in image quality.
Figure 4 shows example results, demonstrating the improved correction capability and field map fidelity of DFD-Net, especially in the posterior regions of the brain. Importantly, as seen in Figure 4(c), the anatomical details are less prevalent in the estimated field map from DFD-Net when compared to that of FD-Net, demonstrating the disentanglement capability of DFD-Net. The disentanglement capability of the proposed DFD-Net provides significant improvements in both image quality and the field map by successfully preventing leakage of anatomical details into the field map. While the supervised baseline provides higher performance for field estimation, it does not generalize well in terms of image correction as it lacks physics-based data consistency. In conclusion, the proposed DFD-Net effectively disentangles image features from the displacement field. It outperforms its predecessor FD-Net in terms of both distortion correction and field fidelity, while providing over 200-fold speedup over TOPUP.
Muhammed Hasan KAYAPINAR (Ankara, Turkey), Abdallah ZAID ALKILANI, Emine Ulku SARITAS
16:06 - 16:08
#47636 - PG200 Time-resolved volumetric speech MRI at 35 frames per second from a one-minute CMR-MOTUS protocol.
PG200 Time-resolved volumetric speech MRI at 35 frames per second from a one-minute CMR-MOTUS protocol.
Speech and related motion patterns are complex dynamic processes in which multiple muscle groups along the upper airways interact to enable vocalization. A clinical example of the importance of imaging speech is cleft palate patients, where MRI has been proposed to assess velopharyngeal function before and after surgery(1,2).
However, the motion involved in speech is challenging to capture by MRI because it generally does not exhibit periodicity and involves non-coplanar motion of the muscles of interest(3,4). (1,2)For this reason, fast MRI sequences to image speech have been typically limited to fast 2D-acquisitions that target the mid-sagittal plane to study velopharyngeal closure and sound production. However, these 2D approaches fail to capture the relevant muscle dynamics/motion outside this plane such as the levator veli palatini muscle. 3D speech imaging approaches have been developed that use non-cartesian (spiral) readouts to volumetrically resolve speech at frame-rates up to 36 frames-per-second. However, these approaches either limit volumetric coverage and resolution, or necessitate a long scan (~10 minutes) during which subjects have to repeat the same phrase, creating practical problems (5–7).
In this work, we show that time-resolved volumetric speech data at 35 frames-per-second and 2-mm isotropic resolution is feasible. Importantly, this is obtained from a one minute long CMR-MOTUS(8) scan implemented with a temporally incoherent pseudo random cartesian acquisition.
We used a pseudo random temporally incoherent Cartesian sampling pattern called PR4D(9,10). This sampling strategy aims to provide uniform coverage in a given time-window. This is achieved by traversing the ky-kz plane incoherently in both the angular and radial direction which yields a sampling pattern that’s a mix between a cartesian spiral and radial filling. We generated a PR4D sampling pattern for a scan with 36000 readout lines. Over the whole scan this yielded a practically fully-sampled k-space with a higher density in the center. An example of a single time-window is shown in Fig 1a while the sampling density is shown in Fig 1b.
A healthy volunteer was scanned using a 3D balanced steady-state free precession (BSSFP) sequence that featured the aforementioned sampling pattern. Imaging parameters were as follows: voxel size = 2 x 2 x 2 mm3, field-of-view = 308 x 220 x 160 (192 with slice-oversampling) mm3 (FH x AP x RL), TR/TE = 2.9/1.43 ms and flip-angle = 50 degrees. During the scan the volunteer was instructed to repeat the days of the week in english. The scans were performed on a 1.5T MRI-scanner (Philips, Best) with a 17-channel head-and-neck receive coil. 20.000 readout lines were used to reconstruct images at a frame-rate of 35 frames-per-second which corresponded to 1 minute of scan time.
The CMR-MOTUS framework was used to reconstruct the time-resolved data. The CMR-MOTUS reconstruction process alternates between image reconstruction step and motion field estimation step to obtain time-resolved images from highly undersampled 3D k-space data(8). Fig 2a shows orthogonal views of single time frame. Here, different structures such as the tongue, velum, and the nasal and oral cavity can be distinguished. A time-series of 30 seconds of fig 2a can be found here: https://surfdrive.surf.nl/files/index.php/s/0iKwm3BUBLQTdPn/download . The volumetric acquisition allowed for the generation of surface renderings which can be seen in Fig 2b while a time-series of 30 seconds can be found in https://surfdrive.surf.nl/files/index.php/s/Fyhftj4o00dnXK5/download
Figure 3 shows a mid-sagittal view and a profile-plot of tongue movement. The mid-sagittal view shows the deformation of the tongue during speech, which can also be observed the 1D projection. A banding artefact can be observed on the velum (blue arrow) which originated from off-resonance during the bssfp acquisition. The presented acquisition scheme showed high spatiotemporal resolution (2 mm/35 fps) with a whole-head coverage. Compared to state-of-the-art(6), the main advantages of our approach is a short scan time of only 1 minute and a larger volumetric coverage which enables the complete visualization of all the surrounding musculature.
The current sequence can still be optimized in terms of sampling and contrast. Currently, the sampling scheme in this work used default setting provided in Joshi et al(10). However, optimization of this sampling scheme in terms of sampling density might yield further improvements in SNR and temporal resolution. In addition, the BSSFP sequence was used to maximize SNR but yielded banding artefacts and low contrast between muscle and other soft-tissue. Alternatively, an RF-spoiled T1-weighted sequence could be used, which would yield more contrast between the tissues at the cost of SNR. This work showed that whole-head time-resolved volumetric speech imaging at 35 fps and 2 mm isotropic resolution is feasible using a 1-minute scan.
Edwin VERSTEEG (Utrecht, The Netherlands), Thomas OLAUSSON, Narjes AHMADIAN, Aebele MINK VAN DER MOLEN, Dennis KLOMP, Cornelis VAN DEN BERG, Alessandro SBRIZZI
16:08 - 16:10
#47411 - PG201 Enhanced Spinal Cord Lesion Detection in Multiple Sclerosis Using White-M atter-Nulled 3D MPRAGE with Deep Learning Reconstruction.
PG201 Enhanced Spinal Cord Lesion Detection in Multiple Sclerosis Using White-M atter-Nulled 3D MPRAGE with Deep Learning Reconstruction.
Multiple sclerosis (MS) is a prevalent inflammatory disease affecting the central nervous system, with spinal cord lesions present in about 80% of cases (1). These lesions contribute to a range of disabling symptoms including sensory loss, motor weakness, and bladder dysfunction (1). Current spinal cord imaging techniques, such as 2D T2-weighted (T2w) and 2D Short Tau Inversion Recovery (STIR) sequences, are commonly used in accordance with the international guidelines (2) but often lack the sensitivity needed to detect all lesions, particularly in the cervical spine. This study evaluates the performance of 3D white-matter-nulled (WMn) magnetization-prepared rapid acquisition gradient echo (MPRAGE) imaging combined with a deep learning reconstruction (DLR) denoising method in improving lesion visibility and detection accuracy while keeping a short scan time.
In this prospective, single-center study approved by the national review board, thirty-eight patients with relapsing-remitting multiple sclerosis (RRMS) or clinically isolated syndrome (CIS) were recruited based on these inclusion criteria: 1) age over 18 years, 2) confirmed RRMS or CIS diagnosis per 2017 McDonald criteria (3), and 3) spinal cord symptoms developed within the last 6 months. MRI exams were conducted using a 3T scanner (Vantage Galan 3T/ZGO, Canon Medical Systems, Tochigi, Japan), involving two joint sessions: one focused on the cervicothoracic spine (C1 to T5-T6) and the other on the thoracolumbar spine (T6 to L2). For both segments, the following sequences were acquired: 2D T2w Fast Spin Echo (FSE), 2D STIR, 3D T1w MPRAGE, and a 3D WMn MPRAGE sequence, which was fine-tuned from an original sequence initially optimized for differentiating thalamic nuclei (4,5). The WMn technique uses an inversion pulse to null white matter longitudinal magnetization while preserving surrounding tissue signals. Due to its short inversion time (470 ms), compared to MPRAGE (950 ms), the 3D WMn sequence has inherently lower SNR, as there's less time for magnetization recovery before reaching steady state. To address this, we consistently applied a manufacturer-provided deep learning-based denoising method.
Four neuroradiologists independently assessed lesions and artifacts from C1 to L2 in randomly ordered cases. Lesion detection confidence was rated as weak, moderate, or strong; artifacts were scored from none to major. A lesion was confirmed if at least 2 of 4 raters had moderate or strong confidence.
Comparisons across sequences included: 1) total lesions per spine level, 2) artifact presence and severity, and 3) lesion contrast-to-noise ratio (CNR). Normality was tested with Shapiro-Wilk, and differences were evaluated using Mann-Whitney U tests (p < 0.05). Analyses were performed in R (v4.3.3). Most lesions were located within the cervical spinal cord (C1–C5). However, a substantial number of thoracic lesions were also observed, accounting for 28% of all lesions detected from T6 to L2 using the 3D WMn sequence (Figure 1A).
Compared to other sequences, the 3D WMn sequence significantly enhanced the detection of lesions across the entire spinal cord (Figure 1A), with particularly notable improvements in the cervicothoracic segment (Figure 1B). In this region, 3D WMn enabled the detection of 62% more lesions than the 2D T2-weighted sequence (p < 0.001), 47% more than 2D STIR (p < 0.05), and 50% more than 3D MPRAGE (p < 0.01). Similarly, in the thoracolumbar segment, 3D WMn outperformed the 2D T2-weighted sequence, identifying 53% more lesions (p < 0.05). It also detected 6% and 25% more lesions than 2D STIR and 3D MPRAGE, respectively, though these differences did not reach statistical significance (Figure 1B).
Figure 2 illustrates the improved lesion delineation in the cervicothoracic region with the WMn sequence. Lesions appeared sharper and more clearly defined, including lesions (arrow) that were missed on the other sequences.
The thoracolumbar images are illustrated in Figure 3 similarly showing superior lesion visualization with 3D WMn. One lesion (arrow) was uniquely detected using this sequence. It also illustrates the higher amount of artefacts with the 2D STIR which impaired lesion visibility despite favorable contrast.
To better understand the improved detection with WMn, we also quantified both artifact prevalence and lesion CNR. The 3D WMn sequence exhibited significantly fewer artifacts than 2D STIR in both spinal segments (Figure 4A). Moreover, it provided a significantly higher lesion CNR than all three other sequences (Figure 4B). 3D WMn MPRAGE combined with DLR is a highly effective imaging technique for detecting spinal cord lesions in MS, offering superior sensitivity due to higher lesion contrast and fewer artifacts compared to conventional MRI sequences. These findings suggest that WMn could play a crucial role in the routine evaluation of spinal cord lesions in MS, potentially improving early diagnosis and treatment outcomes.
Fanny MUNSCH (Bordeaaux), Amaury RAVACHE, Takayuki YAMAMOTO, Bei ZHANG, Marion LACOSTE, Hikaru FUKUTOMI, Pauline BUISSONNIERE, Aurelie RUET, Jean-Christophe OUALLET, Thomas TOURDIAS, Vincent DOUSSET
16:10 - 16:12
#47658 - PG202 DL-QRAGE – Model-based Self-Supervised Physics-Informed Neural Reconstruction for Fast Quantitative MRI of Water Content, T1, T2* and Magnetic Susceptibility at 7T.
PG202 DL-QRAGE – Model-based Self-Supervised Physics-Informed Neural Reconstruction for Fast Quantitative MRI of Water Content, T1, T2* and Magnetic Susceptibility at 7T.
Clinical adoption of quantitative MRI (qMRI) is hindered by lengthy acquisition and reconstruction times. The QRAGE imaging sequence, a multi-echo MPnRAGE-like sequence, acquires over a hundred contrast images at varying inversion and echo times [1]. It produces quantitative parameter maps of water content, T1, T2*, and magnetic susceptibility, achieving full brain coverage with 1 mm isotropic resolution in an acquisition time of about 7 minutes. This efficiency is achieved through a high acceleration factor of R=32. The missing k-space information is then reconstructed jointly, leveraging prior knowledge of the spatiotemporal signal evolution. However, reconstruction times can extend to several hours, posing a significant obstacle to clinical adoption.
Recent advancements in self-supervised deep learning (SSL) have shown promise in reducing MRI reconstruction times from hours to seconds while maintaining or even enhancing image quality [2]. However, existing SSL methods are tailored for conventional MRI and do not fully exploit the unique characteristics of qMRI.
To address this gap, DL-QRAGE is introduced, which integrates self-supervised deep learning with a physics-informed loss function specifically designed for the QRAGE sequence. The network architecture (Figure 1A) is based on the conventional SSL architecture. Unlike conventional SSL reconstruction, the QRAGE network processes multi-contrast data as both input and output and employs a large number of intermediate channels. Additionally, it uses complex-valued convolutions and ReLUs [3].
For network training (Figure 1B), the conventional SSL approach is employed by randomly dividing the k-space data into two disjoint sets. Similar to dual-domain learning [4], DL-QRAGE is run on both sets simultaneously with the same weights, predicting the data from the other set (k-space loss) while ensuring that the reconstructed images are similar (image-space loss). Furthermore, block-Hankel matrices are constructed from the reconstructed time series, and the sum of their nuclear norms is penalized to minimize the number of exponential terms in each voxel (physics-based loss). As an ablation experiment, the model is trained using only k-space loss, with k-space and physics-based loss, and with all three loss terms.
DL-QRAGE is compared to a complex-valued SSL model [2], [3], where each contrast image is reconstructed individually using a different set of weights. Hereby, all SSL models together are approximately the same size as the DL-QRAGE model. DL-QRAGE is further compared to the conventional QRAGE reconstruction.
All data were acquired using a commercial 7T scanner (MAGNETOM Terra, Siemens Healthineers, Erlangen, Germany). QRAGE training data were acquired from 4 healthy subjects with an acceleration factor of R=32. QRAGE inference data were acquired from 4 different healthy subjects using an acceleration factor of R=16 and were retrospectively undersampled to R=32.
Model training and inference were performed at the Jülich Supercomputing Centre, using compute nodes equipped with 4x NVIDIA A100 GPUs. Parametric maps were computed from contrast images via non-linear fitting, and UNI images were created from virtual contrast images [5].
To quantitatively assess reconstruction quality, the mean absolute error is computed between SSL/DL-QRAGE and QRAGE with R=32/16 spokes, respectively. Compared to QRAGE, SSL and DL-QRAGE significantly reduce reconstruction time from 6-8 hours to under 20 seconds. Contrast images and UNI images from all reconstruction methods are shown in Figure 2, while parametric maps are displayed in Figure 3. The mean absolute error for all methods is presented in Table 1 for contrast images, UNI images, and parametric maps. Clearly, DL-QRAGE consistently and significantly outperforms SSL. Incorporating the Hankel loss term (P) improves image quality in nearly all cases compared to using only k-space loss (K). Additionally, including image-space loss (I) yields the best results, with mean absolute errors often closer to the R=16 QRAGE reconstruction than the R=32 QRAGE reconstruction. While DL-QRAGE inference is computationally efficient, training remains resource-intensive due to the model's large size and substantial GPU memory requirements. Additionally, the model is challenging to train and requires small learning rates to avoid instability. Consequently, training a single model currently takes several weeks and improving training efficiency remains a key challenge. DL-QRAGE maintains QRAGE reconstruction quality while significantly reducing reconstruction time and computational resources. This efficiency enhances the clinical adoption of qMRI, enabling timely and accurate disease diagnosis and monitoring. By addressing a major bottleneck in advanced MRI techniques, DL-QRAGE highlights the potential of combining self-supervised deep learning with physics-informed loss functions to advance medical imaging technologies and patient care.
Markus ZIMMERMANN (Jülich, Germany), Felix LANDMEYER, Jörg FELDER, Jürgen DAMMERS, Sohel HERFF, N. Jon SHAH
16:12 - 16:14
#47571 - PG203 Self-supervised deep learning based spectrum denoising in hyperpolarized 13C mri.
PG203 Self-supervised deep learning based spectrum denoising in hyperpolarized 13C mri.
Hyperpolarized 13C MRI (HP MRI) enables metabolic imaging but struggles with low signal strength and noise susceptibility [1,2,3]. Traditional denoising techniques, such as Singular Vector Decomposition (SVD) improve data quality but often fail at low Signal-to-noise-ratio (SNR) and require specific user input [4]. To mitigate this, we implemented a self-supervised deep learning approach and compared its performance to SVD-based denoising [4,5].
We adapted a self-supervised UNet denoising technique for HP MRI that has shown promising results in Raman spectroscopy and has no need for ground-truth data during training [5]. Performance was compared to SVD through simulations and real-world acquisitions. Simulated HP MRI spectra were generated by modeling free induction decay (FID) with added Gaussian noise to assess denoising accuracy.
Simulation of HP MRI Spectra:
We evaluated denoising performance using simulated FID signals generated from predefined metabolite-like parameters. Two sets of simulation experiments were conducted: a single-peak scenario consisting of peak A and a two-peak scenario (peak A and B). In the first scenario, signals were generated with an initial amplitude of 1.0, a T2 decay constant of 0.10 seconds, and a frequency of 10 Hz. The second scenario included an additional second peak B (initial amplitude 0.30, T2 = 0.15 s, frequency 20 Hz). Time-domain signals of 256 samples were acquired uniformly over 1.28 s. To simulate longitudinal relaxation (T1), two successive acquisitions (TP 0 and TP 1) were generated with a spacing of TR = 0.20 s, using T1 values of 1.0 s and 0.8 s for peaks A and B, respectively. The second timepoint was only necessary for SVD-based denoising, which requires a multidimensional input dataset, whereas UNet processes spectra independently. Performance of both methods was assessed at the first timepoint. Complex Gaussian noise was added at three levels, corresponding to peak-SNR (pSNR) ranges of roughly 5–9 (low noise), 2–4 (medium noise), and 1–3 (high noise) to reflect a range of possible pSNR conditions found in experimental HP MRI data (Figure 1A, C). Spectra were obtained via Fast Fourier Transform masked to 0–100 Hz, and noise was quantified in a peak-free band (50–99 Hz). pSNR was computed as the mean amplitude over the peak and its adjacent frequencies (spanning the FWHM), divided by the standard deviation of the noise band. For each scenario, we generated 5 independent simulations per noise level, each followed by 5 denoising trials, resulting in 25 replicates per noise level.
Real World Data:
In vivo data from a healthy mouse brain were acquired on a 9.4 Tesla Bruker preclinical MR system equipped with an RF cryocoil combined with a 1H volume coil, (FID, axial slice, slice thickness: 5mm, acquisition bandwidth: 5000 Hz, number of spectral samples: 8192, repetitions: 24, repetition time: 5000ms, flip angle: 20°). The mouse was injected with hyperpolarized [1-13C]pyruvate produced by POLARIS, a new commercially available preclinical PHIP-based hyperpolarizer. The self-supervised deep-learning-based approach achieved a visible reduction in noise compared to noisy data and SVD in single- and two-peak scenarios (Figure 1). These findings were consistent across different noise levels with high mean pSNR improvements in high and medium noise cases (Figure 2). In scenarios involving single peaks, UNet performed reliably at high and medium pSNR but experienced noticeable degradation at lower pSNR levels, eventually failing below a certain threshold. Additionally, the UNet demonstrated improved denoising performance in the multi-peak scenario when a second high pSNR peak was present compared to the single peak scenario at medium noise levels (Figure 3). Evaluation on in vivo UCSF datasets showcased that UNet visibly suppressed noise, while restoring low-amplitude peaks, such as alanine and bicarbonate, which were difficult to distinguish in the noisy spectra (Figure 4). Compared to SVD-based denoising, the UNet approach demonstrated superior noise suppression. In addition to that, we observed an increased performance in low pSNR regions when high pSNR peak information was present. This is especially important for HP MRI, where metabolites exhibit varying peak intensities. Additionally, the proposed method is expandable, offering further potential for optimization, for example by including information from multiple repetitions. The presented enhancements make this deep-learning-based denoising approach a strong candidate for HP MRI. Future work will optimize models, accelerate inference, and explore clinical integration.
Konstantin MÜLLER (Ulm, Germany), Tamara VASILKOVSKA, Xiao GAO, Meetu WADHWA, Galen REED, Myriam CHAUMEL, Renuka SRIRAM, Jeremy GORDON, Ilai SCHWARTZ, Michael GÖTZ, Pascal P.r. RUETTEN
16:14 - 16:16
#45694 - PG204 SEMAC Ripple Artifact Reduction using Wavelet Domain Filtering for MR Images of Total Hip Arthroplasty.
PG204 SEMAC Ripple Artifact Reduction using Wavelet Domain Filtering for MR Images of Total Hip Arthroplasty.
Slice encoding for metal artifact correction (SEMAC) is often used for MR imaging of total hip arthroplasty to reduce metal artifacts [1]. Spectral profile combination in image reconstruction can create ripple artifacts, impacting diagnoses near implants [2,3]. While slice/bin overlap can fix these artifacts, it substantially increases acquisition time and SAR [4]. Using a B_0-field map may reduce ripple artifacts but also extends acquisition time and can leave residual artifacts [5]. Both methods are impractical in clinical settings. To address this, we developed a wavelet domain filter (WD-Filter) to reduce artifacts retrospectively without prolonging acquisition time.
Wavelet Domain Filter:
Ripple artifacts as they typically occur in SEMAC exhibit a specific distinct distribution in the frequency domain (~0.5-2.0 cm^(-1)) and have a wave-like pattern in the image domain [6]. The wavelet transform (WT) provides frequency and temporal resolution, capturing frequency and spatial information [7]. The Fejér-Korovkin wavelet family with 6 or 12 vanishing moments (fk6 or fk12) shows high absolute values in the detail coefficients for ripple artifacts. Within the ROI, containing a ripple artifact, the detail coefficients were nulled using a threshold level between 12% and 45% of the peak signal. The filtering process for ripple artifact reduction is illustrated in Figure 1A. Additionally, two decomposition levels are employed to address the visible ripple artifacts in the approximation plane. The filter algorithm was implemented in MATLAB R2023b (Mathworks, Natick, MA, USA).
Patient Selection / MR Imaging:
In a retrospective study we included 100 patients with primary hip implants to validate the potential of the WD-Filter. The images were obtained from two 1.5T MRI scanners (MAGNETOM Sola; MAGNETOM Avanto Fit; Siemens Healthineers AG, Forchheim, Germany) at Balgrist University Hospital, Zurich, Switzerland between December 2023 and June 2024. A 32-channel spine-coil combined with an 18-channel surface-coil was used, and images were acquired with a coronal compressed sensing-based STIR SEMAC sequence (TR/TE: 5000/37ms; voxel size: 1.0x1.0mm2; slices: 28; slice thickness: 3.5mm; SEMAC spectral encoding steps: 12 or 19) [8].
Image Analysis:
Two musculoskeletal fellowship-trained radiologists (R1, R2) independently evaluated the ripple artifacts by comparing the ROI of the original images to the images with the WD-Filter. They assessed the intensity of ripple artifacts using a 4-point Likert scale (Likert-RA) with the following scores: 1 = none, 2 = mild, 3 = moderate, and 4 = severe (Figure 1B). Additionally, they measured the standard deviation in a ROI containing a ripple artifact (SD_ROI) before and after applying the WD-Filter. A 4-point Likert scale (Likert-IQ) was used to evaluate image quality improvement, with scores of 1 = poor, 2 = moderate, 3 = good, and 4 = excellent.
Statistics:
A p-value of less than 0.05 was considered statistically significant [9]. The inter-reader agreement for Likert-RA and Likert-IQ scores was assessed using kappa statistics (Cohen's κ) as reported by Landis and Koch [10]. Ripple artifacts were detected in 66 out of 100 patients. We applied the WD-Filter for a total of 250 ripple artifacts.
The Likert-RA improved significantly when the WD-Filter was applied, with an ‘almost perfect’ inter-rater agreement, and there was a significant reduction of 19% in SD_ROI. The Likert-IQ improved significantly with an ‘almost perfect’ inter-rater agreement. The evaluation of both readers for Likert-RA, SD_ROI and Likert-IQ is summarized in Figure 2.
In 59 (R1) and 56 (R2) instances, the original images, initially rated with a Likert-RA score of 4 (severe), were improved to a Likert-RA score of 1 (none) after applying the WD-Filter. Additionally, in 43 (R1) and 47 cases (R2), the original images, initially rated with a Likert-IQ score of 1 (poor), were enhanced to a Likert-IQ score of 4 (excellent). An example of these improvements in both Likert scales is shown in Figure 3.
Furthermore, multiple ROIs in the WD-Filter could reduce more than one ripple artifact in one slice. Figure 4 illustrates an example of a significant improvement in the Likert-RA and Likert-IQ values for two ripple artifacts. The WD-Filter effectively reduced ripple artifacts and significantly improved the overall image quality. However, in 6% of cases, images only achieved a 'moderate' image quality due to residual artifacts, indicating that the filter may not fully address certain shapes or orientations of ripple artifacts. Adjusting the filter threshold level or considering a different wavelet could mitigate this issue. The proposed WD-Filter reduced ripple artifacts and enhanced image quality. The ability to apply the WD-Filter retrospectively on images makes it easy to implement without any need for MRI protocol changes or sequence modifications.
Jeanette Carmen DECK (Zurich, Switzerland), Sophia Samira GOLLER, Georg Wilhelm KAJDI, Constantin VON DEUSTER, Reto SUTTER
|
Salle Major |
|
"Friday 10 October"
Added to your list of favorites
Deleted from your list of favorites
D25
15:40 - 17:10
_______
|
Salle 120 |
|
"Friday 10 October"
Added to your list of favorites
Deleted from your list of favorites
E25
15:40 - 17:10
Medical Device Regulation
|
Salle 76 |
|
"Friday 10 October"
Added to your list of favorites
Deleted from your list of favorites
G25
15:40 - 17:10
Poster 5
FT3 - Ultra-low & Ultra-high field | FT3 - Artifacts and imperfections | FT1 - Technologies for motion
15:40 - 17:10
#45946 - PG411 Open-source quality control procedures for low-field MRI.
PG411 Open-source quality control procedures for low-field MRI.
Quality control (QC) procedures in medical imaging are mandatory[1-3] to achieve reproducible results, which become even more important when the system hard- and software may change such as in open-source low field MRI developments of the OSI² ONE MRI scanner[4]. QC frameworks should be easy to employ, allow for longitudinal comparisons and should be sensitive to hardware, software and environmental changes. QC may differ between initial extensive protocols, fast protocols for routine QC and sophisticated protocols to target specific metrics. We here present a routine QC protocol which helps with the developments of a low field MRI system and comparability/reproducibility of different low-field MRI scanners and is sensitive to spatial as well as temporal system instabilities which may infer image quality.
Open-source QC procedures were implemented including a 3D printed “Hello World” phantom (deionized water with 1.5g/L CuSO4), a reproducible positioning system of the phantom and RF coil, pulse sequences and post-processing routines.[5, 6] Imaging was performed at the 47.5mT OSI2 ONE v1[4, 8] (PTB, Berlin, Germany) including RF coil load tuning (<-20dB) and static linear shimming with calibrated gradient offsets.[8] A 3D turbo-spin echo sequence was developed in PyPulseq[7], imaging parameters are given in Table 1. The read-out (RO) direction was along B0. Phantom data was acquired for 12 consecutive repetitions. Additionally, a volume without RF excitation was acquired for noise measurement.
Images were reconstructed using 3D Fourier transformation. QC metrics were derived from a homogenous coronal slice using an eroded phantom mask. Image quality was assessed by spatial SNR (ratio of mean phantom signal and standard deviation of noise measurement) and temporal SNR (ratio of voxel-wise mean and standard deviation over repetitions). Spatial fidelity was assessed by signal uniformity inside the phantom (range normalized by mean). Larmor frequency drifts during repeated QA scans were assessed using the phantom position within the image (frequency offset causes image shift). In addition, Larmor frequency was measured before and after QC. Figure 1 shows the experimental setup of the low-field MR scanner, including the positioning system to ensure iso-center positioning with high repeatability. Moreover, three planes of the phantom based on QC 3D-TSE sequence are shown. The coronal slice demonstrates the printed features of the phantom including axis asymmetry and a reference point on the right side.
In Figure 2, the visual results of the quality control protocol are shown. The mean image indicated signal inhomogeneity along the RO direction while temporal variations were strongest at the phantom periphery (temporal SNR = 15.8). The automatically determined phantom center (blue) showed a slight off-center position (red). Two distinct electromagnetic interferences were visible in the scan, which increased the overall noise level (mean=6.1, std=3.5).
Figure 3 presents QC metrics over repetitions. Spatial SNR (mean=19.5) decreased over time whereas signal uniformity (mean=39.3%) did not show a trend. An increasing off-center shift along the RO direction (0.18px/repetition) was measured but not along phase-encoding directions. The Larmor frequency was increased by 720 Hz after QC scans. We present a QC protocol to monitor the performance of low-field MR systems. QC data can be collected frequently to assess the impact of hardware or software modifications or to optimize MR sequences. The phantom, positioning system, sequences, raw data and analysis pipeline are made available open-source.[5, 9]
The QC analysis revealed an inhomogeneous signal distribution along the RO direction which could be due to the narrowband tuning of the RF coil. The linear regression revealed a shift of the phantom possibly due to Larmor frequency drifts induced by temperature changes. This is particularly relevant for signal averaging typically used to improve SNR in in-vivo imaging. The noise scan showed RF interferences which were also visible in the phantom data. As the MR scanner was operated outside an RF shielded room, the sources still need to be identified; SNR decreased across repetitions which could be due to a shifted Larmor frequency in conjunction with the narrowband tuning of the RF coil. A wideband tuning of the RF coil or an automatic coil matching/tuning to an updated Larmor frequency could resolve the issue. In conclusion, our results demonstrated the importance of a QC protocol for low-field MR development. Based on a few metrics, we identified important system characteristics. In the future we will be able to track better the influence of system modifications or environmental changes on the system’s performance. Sharing not only the QC sequences but also the phantom design should facilitate QC comparisons across different systems and enlarge the library of available QC procedure for dedicated system performance monitoring.
Helge HERTHUM (Berlin, Germany), David SCHOTE, Ilia KULIKOV, Frintz JAN GREGOR, Tobias MOHR, Sebastian SCHACHEL, Christian ENGLER, Stefan HETZER, Christoph KOLBITSCH, Lukas WINTER
15:40 - 17:10
#45623 - PG412 Accelerated Combined caLculation of Ultra-high field Biases (CLUB) with Sandwich: 3D, fast, simultaneous mapping of B0 and B1+ inhomogeneities at 7T.
PG412 Accelerated Combined caLculation of Ultra-high field Biases (CLUB) with Sandwich: 3D, fast, simultaneous mapping of B0 and B1+ inhomogeneities at 7T.
While ultra-high field (UHF) MRI offers improved signal-to-noise ratio compared to lower field strengths [1], its inherently stronger B0 and B1+ field inhomogeneities can result in signal dropouts [2]. Although solutions, such as parallel transmission (pTx) [3], have been developed to mitigate these inhomogeneities, online mapping of B0 and channel-wise B1+ fields is often required.
To reduce this additional scan time, a rapid 3D method, CLUB-Sandwich [4,5], was recently developed for simultaneous B0 and B1+ mapping at UHF, and deep learning (DL) image reconstructions were explored to accelerate B1+ mapping [6].
In this work, we investigate accelerating the CLUB-Sandwich sequence using a DL reconstruction to acquire accurate ΔB0 and multi-channel B1+ maps in under 10s.
Study Population and Image Acquisition
Ten healthy volunteers (five female, age range=[25-41]y/o) were scanned at 7T (MAGNETOM Terra.X, Siemens Healthineers, Forchheim, Germany) with an 8Tx/32Rx head coil (Nova Medical, Wilmington, USA). For each subject, a 3D coil-cycled multi-channel fully sampled acquisition of relative B1+ maps followed by a fully sampled 3D CLUB-Sandwich research application sequence for simultaneous B0 and absolute B1+ mapping was acquired (see Fig.1 for sequence diagram and parameters). In 3 subjects, the CLUB-Sandwich sequence was also prospectively accelerated (R=4 and R=8) using Poisson disk undersampling [7]. An MP2RAGE sequence [8,9] was acquired as anatomical reference.
Image Reconstruction and ΔB0 and B1+ Mapping
The fully sampled datasets were retrospectively undersampled using Poisson disk masks with different accelerations (R=2, 4, 6, 8, 10, 12). Data were then reconstructed using two methods: a joint transmit low rank reconstruction algorithm (TxLR) [10,11] and a DL approach [12], which were compared to the fully sampled reference data. Reconstruction times were also compared to investigate feasibility of online implementation. The performance of the proposed DL approach was further validated on the prospectively acquired data.
The TxLR method enables joint reconstruction of undersampled data using structured low-rank tensor completion [11]. To exploit redundancy across contrasts, pseudo-complementary masks were generated by modifying the seed of the Poisson disk generator, as optimized in previously validated methods [10].
The DL reconstruction, based on variational networks [13], reconstructs 3D images from undersampled k-space data through 6 iterations of data consistency updates and network-based image regularization. The model was pre-trained using 5000 pairs derived from fully-sampled 3D datasets from healthy subjects scanned at 1.5 and 3T, and fine-tuned for 7T using 1000 pairs [12].
ΔB0, absolute, and channel-wise B1+ maps were then computed from the reconstructed contrasts, following the same procedure reported in previous studies[5].
Image Analysis
ΔB0 and B1+ accelerated maps from CLUB-Sandwich data were compared to the fully sampled reference by analyzing the volumetric root mean squared error (vRMSE) in brain tissues segmented from the MP2RAGE datasets using a research application software[9,14,15]. Fig.2 shows ΔB0 and absolute B1+ maps for a representative subject at a retrospective acceleration of R=8, reconstructed using both TxLR and DL algorithms. The corresponding differences to the fully sampled reference demonstrate similar vRMSE. TxLR exhibits oversmoothing in regions with stronger inhomogeneities, while the DL approach yields slightly noisier estimates.
vRMSE values for retrospective data across all subjects as a function of R are shown in Fig.4. DL achieves comparable performance to TxLR for ΔB0, absolute and relative B1+ estimation. Although TxLR presents marginally better accuracy in all three cases, DL requires only 5s of reconstruction time compared to the 4 minutes required by the TxLR.
Prospective DL acceleration results for an illustrative subject are shown in Fig.5. Across the 3 prospective datasets, vRMSE in the absolute maps were found to be 3.2±0.8 Hz and 2.0±0.8° for R=4, and 4.3±0.9 Hz and 2.8±0.9° for R=8, consistent with retrospective results. The proposed DL reconstruction enables accurate acceleration of the CLUB-Sandwich method, offering rapid simultaneous ΔB0 and B1+ mapping with performance comparable to fully sampled data and previously validated techniques [10]. By reducing acquisition and reconstruction times to 9s and 5s, respectively, the approach supports real-time implementation at the scanner, enhancing efficiency of UHF workflows. Prospective results validate the ability to generate reliable B0 and B1+ maps in under 10s. This work validates a DL reconstruction approach for accelerating inhomogeneity mapping at UHF, maintaining accuracy while reducing scan time. Future work will focus on demonstrating the reliability of the estimated maps for online in vivo pTx optimization at 7T and exploring the impact of a DL-based joint reconstruction across the acquired contrasts.
Natalia PATO MONTEMAYOR (Lausanne, Switzerland), Jocelyn PHILIPPE, James L. KENT, Aaron HESS, Antoine KLAUSER, Emilie SLEIGHT, Lina BACHA, Tommaso DI NOTO, Bénédicte MARÉCHAL, Patrick A. LIEBIG, Juergen HERRLER, Dominik NICKEL, Robin M. HEIDEMANN, Tobias KOBER, Jean-Philippe THIRAN, Tom HILBERT, Thomas YU, Gian Franco PIREDDA
15:40 - 17:10
#47639 - PG413 Spatial-spectral parallel transmission RF pulses design for adiabatic spin inversion.
PG413 Spatial-spectral parallel transmission RF pulses design for adiabatic spin inversion.
In NMR and MRI, adiabatic RF pulses – characterized by a linear RF frequency sweep – improve drastically tolerance to heterogeneous B0 and B1 fields over ordinary linear phase pulses [1]. Parallel transmission (pTx) is a recent evolution of the RF hardware of MRI where RF transmission is accomplished with an array of resonators driven independently over time [2]. Owning to Pauly’s concept where RF and MFG shape are applied in concert to control spatially the spin system’s excitation profile [3,4], pTx offers a practical (short excitations) low-SAR alternative to frequency sweeping. Nevertheless, Pauly’s spatial pulses on their own do not replace at all adiabatic pulses because they lose in general a clean spatially invariant spectral definition, a prerequisite in many applications, in particular, slice selection. Recently, a method to create 3D composite adiabatic pulses exploiting the full pTx capability of the system was described [5,6]. In essence, this method is a pTx variant of the spatial-spectral pule design concept [7]. They first choose the so-called parent pulse defining the desired spectral profile, e.g the hyperbolic-secant adiabatic (HS) pulse. Second, they compute a short 2D-selective pTx spatial pulse using the spatial domain method [8]. Third, they form a train of the latter pulse and modulate the RF amplitude of each Tx channel using the parent pulse’s RF shape. This technique was shown to enable B1 and B0 insensitive 3D-selective inversion in a short time. In this work, we revisit this approach with a HS parent pulse (bandwidth B) and a short (T=200us, BT<<1) non-selective pTx “modulating” pulse yielding here a uniform flip angle profile. We show that the composed pulse continues to behave like an HS (the parent shape) with the advantage that the modulating subpulse promotes the fulfillment of the adiabatic condition [1] uniformly across space. Under difficult experimental conditions – particularly, in this work the absence of circular symmetry in the Tx array arrangement – where static RF shimming [9,10] becomes inoperant to maintain the adiabaticity condition across the entire volume of interest, the proposed approach appears constructive, and potentially very useful.
Let w be the parent waveform and p=(g,v) be a candidate spatially uniform 1° non-selective spatial pTX pulse of duration τ (g is the B0 gradient shapes in x, y and z and v is the RF shapes on each TX channel), with g self-refocused (i.e. ∫g(t)dt=0). Let Cτ be the τ–periodic Dirac comb distribution. We define the product pulse as:
W∶=(Cτ ∗ g, λ(Cτ w) ∗ v) (Eq. 1)
with ∗ the convolution product and λ an adjustable positive RF scaling factor. We can show that this so-called product pTx pulse performs uniformly across space, and frequencies within the [-0.5/τ,0.5/τ] frequency band. We note that the ∫g dt =0 condition is important to ensure that the propagator of p [11] has a transverse rotation axis.
An experimental validation of this pulse design approach was performed on a Magnetom 7T MRI (SC72, 8x1kW), using the home-made “Avanti2” 8Tx32Rx head coil prototype [12]. For the parent pulse, we used a 10-ms HS inversion pulse with β=5 and B=1kHz. Measurements were performed on 16 cm-diameter spherical phantom filled with agarose (εr=72, σ=0.8S/m). The B0 and B1+ measurements were performed using a multi-echo 3D GRE and an interferometric 2D SatTFL acquisition (5 mm isotropic resolution) [13]. The 8 B1+ profiles, the combine mode (CM) B1+ profile, and the RMS B1+ profile are displayed in Fig.1. A 1°, 200µs spatial subpulse was designed using fastGRAPE [14] (Fig.2a). Application of Eq.1 (λ adjusted to yield the same peak amplitude as the parent shape) produced the product pTx HS pulse shown in Fig.2b. Finally, a preliminary validation of this approach was performed with a 3D MP2RAGE acquisition (TR/TI1/TI22=3.3/0.83/2.5 s, FA1/FA2=4/2 deg). The pTx modulating pulse yielded a flip angle normalized RMS error (NRMSE) of 16.2%. Retrospective flip angle simulations for the pTx modulating pulse, the CM-HS pulse (NRMSE = 17.3%, energy=8.7J), the product pTx HS pulse (NRMSE=5.2%, energy=9J) and two slice selective (3cm and 1cm slice thickness) derivatives of the latter are shown in Fig. 3. While the HS pulse fails to invert the magnetization in the regions indicated in Fig.1, the product pulse’s inversion efficiency is uniformly high across the entire object. Slice selection is feasible using the product pulse for the 3cm but not the 1cm case due to the 1/200us cutoff frequency, leading to slice replicas in 5cm interval in this case. We have shown a methodology to exploit dynamic pTx to leverage the performance of the hyperbolic secant adiabatic pulse in the presence of heavily non-uniform B1 profiles. This strategy could be important for imaging at ultra-high fields were standing wave effect can hinder many applications [15,16].
Vincent GRAS (France), Natalia DUDYSHEVA, Franck MAUCONDUIT
15:40 - 17:10
#47030 - PG414 Experimental validation of a simulation model for minimizing eddy-current-induced fields in low-field MRI.
PG414 Experimental validation of a simulation model for minimizing eddy-current-induced fields in low-field MRI.
Low-field MRI systems are especially vulnerable to electromagnetic interference, reducing image quality and signal-to-noise ratio (SNR). Conductive shieldings mitigate these effects but also induce eddy currents that distort the magnetic field [1]. In portable MRI systems [2], these issues are amplified by operation in uncontrolled environments and the use of lightweight, low-cost structures. Understanding how surrounding metal affects the field in the field of view (FOV) is thus key for the design of mechanical structures in MRI scanners. This study presents a frequency-domain simulation framework, which we have validated with controlled experiments.
Eddy currents were studied using a frequency-domain FEM model in COMSOL. While time-domain simulations can reproduce realistic transient signals, they are computationally expensive. Some frequency-domain methods simulate multiple spectral components via Fourier decomposition of the gradient waveform [3] but require fine meshes to capture high frequencies [4]. Instead, we use a single fixed frequency, set as the inverse of the gradient fall time, to capture trends and enable fast parametric sweeps.
While other approaches aim to generally reduce eddy current density in nearby conductors [5], ours targets B_eddy directly in the FOV and focuses exclusively on suppressing the relevant components, i.e. along B₀. This gives us an effective basis for optimization.
The model was validated by placing the scanner magnet and gradient coils in an open environment, free from metallic structures. Aluminum plates in various shapes (solid and frame-like) were placed around to induce eddy currents in a controlled manner (Figure 1). To simplify comparison with simulations, the field was evaluated at the isocenter, where the gradient field is zero, so only eddy-induced fields are present. To replicate this condition, a one-loop circular RF coil (1 cm diameter) was used to confine the signal acquisition region and minimize spatial-integration effects.
To measure the B_eddy, a 10 mT/m gradient pulse was applied, with a 400 µs ramp-down preceding signal acquisition. The field was retrieved from the phase difference between positive (ϕ+(t)) and negative (ϕ-(t)) gradient polarities [6]:
B_eddy (t)=(1/γ) * d/dt ((ϕ+(t)-ϕ-(t))/2)
Simulated values were compared to the value of the B_eddy curve just after the ramp down (t=0). Only the component along B₀ was considered in the simulations, as it is the only that affects the free-induction-decay (FID) signal.
After validation, the model was extended to simulate the field across the FOV. Eddy-induced contributions were isolated by subtracting a baseline simulation, without metallic elements, from the ones including them. This enabled spatial mapping of B_eddy for identification of geometries that minimized eddy effects in the entire FOV. Figure 2 shows numerical and experimental results for an aluminum plate displaced along the X-axis (Figure 1b). Simulations predicted the worst position at a 100 mm offset for the X-gradient and a doubling of the induced field when a second plate was added symmetrically (Figure 1c). For the Y-gradient, both simulations and experiments showed negligible effects. They also correctly predicted the worst case for the Z-gradient, with the plate at the center.
Figure 3 shows the spatial variation of eddy currents across the FOV for two plates displaced along X in the Z-gradient case. Figure 4 shows the numerical results of the FOV-averaged B_eddy for that case. Simulations with metallic plates displaced along different axes correctly predicted the worst positions, i.e. those that caused the strongest field perturbations at the magnet center. Simulated and measured values across different scenarios are repeatedly consistent up to a constant factor. This repeatability is the relevant metric to benchmark the performance of our approach, not its magnitude, as we simulate a continuous sinusoidal excitation rather than the applied trapezoidal pulse.
Finally, extending the analysis to the full FOV is essential, since relying only on the central field may lead to the false conclusion that eddy currents cancel out. This can be observed in Figure 4 for the case of two plates, where the field is nulled only at the center. A frequency-domain FEM model was validated experimentally for evaluating eddy currents in low-field MRI. In all cases, it identified which geometric configurations produced the strongest fields at the scanner center. Its computational efficiency enables rapid parametric studies. By extending the analysis across the full FOV, the method provides insight into how surrounding metallic components influence the B₀-oriented eddy fields, enabling design optimization explicitly aimed at preserving image quality.
Lorena VEGA-CID (Valencia, Spain), Marina FERNÁNDEZ GARCÍA, Jose BORREGUERO, Teresa GUALLART-NAVAL, Eduardo PALLÁS, Lucas SWISTUNOW, Jose Miguel ALGARÍN, Fernando GALVE, Joseba ALONSO
15:40 - 17:10
#45919 - PG415 Introducing a model for predicting acoustic energy in EPI and its correlation to ghost correction at 7T and 10.5T.
PG415 Introducing a model for predicting acoustic energy in EPI and its correlation to ghost correction at 7T and 10.5T.
Echo-Planar Imaging (EPI) is the workhorse of functional MRI. It drives the system aggressively, quickly alternating the currents through the gradient coils. Alternating gradients lead to mechanical vibrations due to the B0 magnetic field applying a Lorentz force. These vibrations produce the well-known MRI sounds, may cause ghosting artifacts, and possibly even mechanical failure. All these are exacerbated at ultra-high fields (as higher B0 means stronger Lorentz forces). New hardware designs aim to reduce vibrations[1-3], but in software, the current approach is to “forbid” certain frequencies, assuming a single driving frequency of one over twice the EPI’s echo-spacing (ESP). In practice, however, multiple audible frequencies arise. In this study, we present a model that predicts these frequencies and suggests that subtle timing changes — of TEs (in case of multi-echo acquisitions) or slices — can affect the acoustic spectrum and imaging ghosts. A procedure to estimate mechanical resonance “amplification” is built, thus estimating also the expected acoustic energy for a specific EPI scan. The combined model can be used to reduce the overall strain on the system. We also examine the correlation between the acoustic characteristics and the ghost correction, which improve our understanding and help to reduce ghost intensities[4-6]. The acoustic energy characteristics and the mechanical resonance “amplification” were studied for 7T and 10.5T scanners, demonstrating the ability to reduce acoustic energy and ghost intensity in both.
The analytic model presented here considers only the echo train readout gradients, modeling them as finite sinusoidal or trapezoidal echo-trains, split between N_"echo" TEs (when relevant) and N_"slice" slices.
To assess the actual effect of timing-changes we implemented an EPI pulse sequence with a per-TR control of slice timing, on/off switching of specific gradients, and an option to shift the navigators timing.
The estimation of the system’s “amplification” included the following key points:
Audio recorded multiple scans with a range of ESPs and time between slices.
Only the readout gradients during the echo trains were on, to best match the (trapezoid-based) model.
Short echo-trains were used to achieve wider spectra.
The “amplification” was calculated as a ratio between the actual FFT of the audio recording and the model spectrum.
Phantoms were scanned on a 7T MRI (Terra, Siemens) with a Nova 1Tx/32Rx coil and on a 10.5T (Siemens) MRI with a 16Tx/80Rx head coil[7]. The same scans were tested in human scanning at 7T. Audio was recorded using OptiSLM 100 (OptoAcoustics) at 7T MRI and with Bruel and Kjaer Type 2237 SPLmeter at 10.5T. Fig. 1A shows the acoustic spectrum squared predicted by the model. Here two ΔTE (timing between echoes) and ΔTslice (timing between slices) cases are shown – i) “arbitrary” (top) and ii) on the “2ESP-raster”, i.e. ΔTE and ΔTslice are multiples of 2ESP. Also shown are the factors whose product makes up the model: the single echo-train contribution (yellow), the multi-echo factor (purple), and the multi-slice factor (green). The waveform on the 2ESP-raster results in a dominant peak at 1/2ESP, instead of several peaks. Fig. 1B shows the estimated mechanical resonance “amplification” measured at 7T with a small surface coil and with the Nova coil, and at 10.5T with a head coil. Three high amplification zones are observed near the system “forbidden frequencies” zones (the Nova coil setup shows higher amplifications). The 7T and 10.5T amplification profiles display certain level of similarity, as expected, since the same gradient coil is employed in both. Fig.2 shows significant changes in the acoustic energy, measured and predicted, as a function of ΔTslice. Two cases are demonstrated - i) ESP=0.53 ms, useful for fast acquisitions, whose first harmonic falls in a high amplification zone, ii) ESP=1.26 ms, useful for high resolution, whose third harmonics falls in a high amplification zone. Fig. 3 shows acoustic energy and ghost intensity as function of navigator and slice timings. Fig. 4 shows sample of in-vivo results. A model-based prediction of the acoustic frequencies in EPI was developed. The model predicts well the acoustic peak locations and shows similar trends of the energy changes as function of timing between slices. We built a method to characterize the mechanical resonance “amplification” of the system, examining it in 7T and 10.5T MRI. The results show that small timing changes (≲2·ESP, few ms) can have significant effects on acoustic energy by up to a factor of ~3 and on ghost level by up to a factor of ~5; exhibiting similar behavior at 7T and 10.5T. The ghost correction in both systems exhibit strong dependence on the timing from the previous acquisition and scan’s acoustic characteristics. Further study of the exact correlation between ghosts and acoustic spectra is required.
Amir SEGINER, Alexander BRATCH, Noam HAREL, Essa YACOUB, Rita SCHMIDT (Rehovot, Israel)
15:40 - 17:10
#47759 - PG416 Improving SNR in areas of high inhomogeneity in Ultra-Low Field MRI using Composite Refocusing Pulses.
PG416 Improving SNR in areas of high inhomogeneity in Ultra-Low Field MRI using Composite Refocusing Pulses.
Ultra-low field (ULF) magnetic resonance imaging (MRI) offers advantages over high field (HF) MRI, including increased accessibility, reduced costs, and portability [1]. However, since MRI signal scales with the magnetic field strength, ULF MRI inherently suffers from reduced signal-to-noise ratio (SNR) compared with HF scanners, thereby limiting its clinical application [2].
The low SNR is reduced further by two additional factors: (1) field strength inhomogeneity (ΔB₀) is significant due to the magnet design in ULF systems; (2) radio frequency (RF) field strength inhomogeneity (ΔB₁) where, for example, a 20% drop is observed at inferior regions of the brain near the edge of the coil.
Composite refocusing pulses (CRPs), consist of a series of contiguous RF pulses with varying amplitudes and/or phases. CRPs have been employed in HF nuclear magnetic resonance (NMR) spectroscopy to mitigate issues related to ΔB₀ and ΔB₁, thereby improving SNR relative to that of simple square pulses.
While CRPs have been used extensively in NMR spectroscopy, their use for in vivo imaging at HF has been restricted due to the high specific absorption rate (SAR) associated with these pulses. At ULF, however, SAR is substantially lower than at HF [3], enabling the potential application of CRPs for enhancing image quality.
Here we consider the 3D Turbo Spin Echo (TSE) sequence - the most commonly used sequence in ULF MRI. Each of the three CRPs shown in Table 1 replaced the square refocusing pulses, and these were compared to square refocusing pulses to assess imaging in both phantom and in vivo scanning in the presence of ΔB₀ and ΔB₁.
Equipment:
Data were acquired on a Multiwave MGNTQ 50mT [8] at CISC in Brighton, UK, and a custom-built 47mT scanner [9] at University of Leiden, NL, both equipped with Halbach permanent magnet arrays and using a solenoid head coil and a Kea2 Magritek spectrometer [10]. In vivo data was acquired on the MGNTQ system.
ΔB₀:
CRPs were implemented in a 1D spin echo pulse sequence, using a NiCl solution (12.5 mL, 10mM). To assess CRP performance in the presence of ΔB₀, off-resonance conditions were created by modulating the centre-frequency of the spectra in the range of ± 2.75kHz in increments of 0.050kHz.
ΔB₁:
To assess the effect of ΔB₁, refocusing pulse amplitudes were modulated from the experimentally-determined optimum for both square pulses and CRPs, in the range B₁/B₁ₒₚₜ=ζ =0.55 to ζ =1.45 in increments of ζ =0.01. The spin echo signal intensity was used to assess the performance of the CRPs compared to that of a square pulse using a solution of water and CuSO₄ of 200mL.
In vivo ΔB₀:
TSE data was collected on the brain of a 42-year-old female. The effect of CRPs was assessed by measuring SNR change, with respect to square refocusing pulses, in two regions-of-interest (ROIs) of low (Fig. 2; ROI1) and high (Fig. 2; ROI2) ΔB₀, respectively. ROIs were identified using a B₀ map (Fig. 2a) co-located with the TSE volumes . To assess overall signal intensity across the entire brain, whole-brain histograms were plotted for each CRP (Fig. 3). A brain extraction tool (BET) [11] with manual cleanup was used. B₁ inhomogeneity: LF and LT exhibited greater insensitivity to ΔB₁ while SP showed little improvement over square (Fig 1a).
B₀ inhomogeneity: In vivo, areas of low ΔB₀ (Fig 2a; ROI1, average ΔB₀ = -36Hz), LT and SP boost SNR by 13% and 11%, respectively, while LF shows a slight reduction in signal (Fig. 2c).
In areas of high ΔB₀ (Fig 2a; ROI2, average ΔB₀=-1700 Hz), both LF and LT boost SNR by over 40% while SP has a smaller but notable improvement (Fig. 2c).
The whole-brain histogram (Fig. 3) showed up to 18% increase in the histogram peak height for all CRPs, which suggests signal inhomogeneity is reduced across the entire brain, presumably due to improved ΔB₀ and ΔB₁ compensation. There was also a shift in the histogram position by 4% for LT, indicating a general increase in voxel intensity across the brain. There was significant increase in signal up to 42% in areas of high ΔB₀ (ROI 2) where CRPs supplanted square refocusing pulses in a TSE sequence.
LT showed the highest SNR increase in both ROI1 and ROI2 (Fig. 2) out of all refocusing pulses, evidenced by the corresponding shift of the whole brain histogram (Fig. 3). This increase could be partially due to LT SNR increase in areas of high ΔB₁. However, more experimentation is needed to fully disentangle the causal factors. This work highlights the potential for boosting SNR in ULF MRI through the use of CRPs.
There is a significant increase in SNR across the whole brain particularly in areas of high ΔB₀. LT showed the largest SNR increase of all refocusing pulses evaluated. This improvement is significant, as CRPs can be incorporated into any sequence which makes use of refocusing pulses and thus it offers the potential for improving the utility of ULF MRI. In future work we plan to investigate CRPs for use with surface coils, which exhibit higher levels of ΔB₁.
Finn AUBREY CONBOY (Brighton, United Kingdom), Samira BOUYAGOUB, Itamar RONEN, Ivor SIMPSON, Chloe NAJAC, Nicholas G DOWELL
15:40 - 17:10
#47951 - PG417 Optimization of tailored ptx spokes pulses for simultaneous multi-slice fmri with pypulseq at uhf.
PG417 Optimization of tailored ptx spokes pulses for simultaneous multi-slice fmri with pypulseq at uhf.
Ultra high-field MRI promises massive improvements in spatiotemporal resolution, but suffers from inherently inhomogeneous B₁⁺ fields. The adaptation of parallel transmit (pTx) technology has long been investigated as effective mitigation of both transmit and main field inhomogeneities. However, in order to leverage these benefits they must be implemented in modern sequences. Simultaneous multi-slice (SMS), or multi-band imaging has been a staple of functional imaging due to the reduced g-factor noise penalty at high acceleration factors. However, combining custom pTx pulse designs with vendor supplied sequences proves difficult due to several technical limitations. To address this issue, we recreate a vendor SMS GRE-EPI sequence in PyPulseq [1,2,3] and implement subject-tailored pTx spokes pulses.
The design of our pulses is based on the spatial domain method with small tip angle approximation [4], solving the magnitude least squares problem in 20 iterations [5]. The k-space trajectory is optimized by sampling a set of up to 10,000 spoke combinations, where the first positions are uniformly sampled from −10 m⁻¹ ≤ kₓᵧ ≤ 10 m⁻¹ and the last spoke is placed at the center of k-space. Pulse energy is controlled via Tikhonov regularization and the best pulse is selected based on minimum normalized root mean square error (nRMSE) of flip angle deviation. In order to enable fast online computation, large batches of spoke combinations are optimized in parallel on a modern GPU.
Due to multi-band pulses being the complex sum of single-band pulses with a frequency offset, the optimization problems for each band may be viewed independently. However, if no regulation is applied, adjacent slices frequently exhibit different inhomogeneity patterns that do not align spatially, leading to zebra stripe artifacts in slice direction. To mitigate this problem, the input slab for the optimization is chosen slightly thicker than the actual excitation, leading to improved stability and continuity at the slab borders.
A blipped-CAIPI [6] sequence with SMS factor 3 and CAIPI shift d_z = 1 was implemented in PyPulseq and the standard excitation pulse replaced by arbitrary pTx multi-band spokes pules. The bandwidth of multi-band pulses is rather high due to the frequency offset between the bands being explicitly encoded as phase evolution. As this may cause aliasing with the default RF raster time of 10 us, the Pulseq interpreter requires minimal modification to accept finer raster times, such as 2 us. Furthermore, the current version v1.5.0 does not officially support pTx pulses, but a convenient hack to the interpreter proposed by Dario Bosch simply adds the channel dimension to pulse waveforms.
As the use of sequence labels in Siemens online reconstruction is vaguely documented at best, a 2D GRAPPA-based [7] offline reconstruction was developed to leverage the a priori information of the sequence such as the CAIPI pattern. Simulations using a database of in vivo B₁⁺ and B₀ maps (n = 14) demonstrated significant improvements in excitation homogeneity with the proposed methods (Figure 1). Compared to CP mode (mean nRMSE = 23.6%), 2-spoke bipolar pTx pulses reduced flip angle error to 13.6% when optimized for 13 mm slabs with 33% overlap. Tailored RF shims and monopolar spokes achieved intermediate performance.
Preliminary results from a single in vivo scan compare CP mode with a single spoke pTx pulse. We map the temporal variability by computing the voxel-wise standard error over nine repetitions after three dummy scans, as shown in Figure 2. While the error is consistent across slices in CP mode, the pTx pulses exhibit some positional dependency with regard to their stability. Parallel transmission offers great advantages and may even be necessary at 9.4 T and beyond for anatomically or functionally demanding regions of interest. Yet technical challenges remain. Subject tailored pulses demand a significantly longer pipelines and may not be worthwhile compared to good universal pulses. The use of custom, open-source sequences however adds flexibility and reproducibility that is invaluable in our opinion. Furthermore, the sequence proved to be more performant that the vendor sequence with room for improvement by implementing ramp sampling, asymmetric spokes or fine-tuning dwell time and spoiling. To ensure compatibility, the aforementioned hacks need to be refined and implemented in the official Pulseq interpreter. This work demonstrates the technical feasibility and performance advantages of using subject-specific, GPU-optimized pTx spokes pulses in a custom SMS GRE-EPI sequence at 9.4 T. While the method shows strong potential in both simulation and initial in vivo results, widespread deployment will require further development of the Pulseq framework and improved hardware interfacing. These efforts are critical for making fast UHF acquisition techniques reproducible, portable, and clinically relevant.
Tim HAIGIS (Tübingen, Germany), Dario BOSCH, Klaus SCHEFFLER
15:40 - 17:10
#47673 - PG418 Computational EM Simulation of Microscopic Graphene-Based Electrophysiology Probes at 7 Tesla MRI: Acceleration Using a Huygens' Box-Based Approach.
PG418 Computational EM Simulation of Microscopic Graphene-Based Electrophysiology Probes at 7 Tesla MRI: Acceleration Using a Huygens' Box-Based Approach.
Concurrent electrophysiological fMRI recordings are a powerful technique that simultaneously records electrical brain activity and hemodynamic changes, providing valuable insights into normal and pathological brain states. This multi-modal technique poses challenges, including metal-based artifacts from conventional probes that distort MRI images. Additionally, RF-induced heating in the vicinity of the electrophysiological recording components. New probes based on Graphene Solution-Gated Field-Effect Transistors (gFET) allow high-fidelity DC-coupled invasive brain signal recordings in rodents [Bonaccini Calia et al, 2022], but their suitability for the MR environment remains to be evaluated. Computational electromagnetic (EM) simulations are useful to evaluate the interactions between implants and the MRI fields. However, due to the microscopic (sub-micrometric) substructures of the gFET probes, the simulation times using standard techniques can be excessively lengthy (>1000 hours). Here we explore the use of a Huygens' Box (HB)-based approach to reduce computation times and improving spatial resolution compared to conventional methods [Neufeld et al, 2009].
EM simulations were performed using the finite-difference time-domain (FDTD) method on a Windows 11 PC (3.00GHz, 32GB RAM, Nvidia RTX 4090 GPU) using the aXware kernel on Sim4Life (V8.0, ZMT, Switzerland; http://www.zurichmedtech.com) in a 3-dimensional (3D) rodent model consisting of 68 tissues [Kainz et al, 2006].
• Simulation setup: 300 MHz of Gaussian excitation with a bandwidth of 625 MHz was used as the excitation source in both methods:
(1) Multi-port (MP) method: Same excitation source in a two-port configuration, followed by impedance matching and combining the results in circular-polarized mode.
(2) Huygens’ box (HB) method: Two-step process: The first simulation is performed with the RF coil excited in circular-polarized mode, generating fields in a rectangular region of interest: the HB. The second simulation uses the HB as a source with the 3D rodent model placed inside the HB; for comparison purposes simulations were performed with and without a probe; see Figure 1.
• RF coil modelling: A quadrature birdcage RF coil is used, with each rung (width: 9.9 mm) containing a capacitor (13.8 pF) placed on the end rings (width: 11.5 mm) to resonate at 300 MHz; see Figure 1.
• Probe modelling: The 3D probe models were generated from 2D drawings and exported to individual layers via Rhino (V8, Washington, DC, USA) and finally converted to multi-layered 3D model in Sim4Life; see Figure 2.
• Estimated EM fields: Transmit RF field (B1+), mass-averaged, and peak spatial-averaged specific absorption rate (SAR) averaged over 0.01 g, 0.1 g, and 1 g tissue mass were calculated following IEC guidelines [IEC]. Table 1 shows the computational times for different simulation types and their respective grid sizes. Note the higher resolution and shorter simulation times for the HB method. The B1+-field distributions in the rodent model for MP and HB simulations were similar, with improved resolution for the latter; B1+ magnitudes in the vicinity of the probes were elevated by approximately 15–20%. Figure 3 shows the SAR distributions in the rodent model for the MP and HB simulations, revealing elevated SAR near the probe. The HB approach enhances electromagnetic simulations by improving computational efficiency and enabling higher-resolution field calculations around probes and the rodent model. However, a limitation is the inability to apply RF coil matching in HB simulations as effectively as in multi-port simulations, introducing slight uncertainties. SAR elevation in the vicinity of the graphene-based probes is modest. This study successfully demonstrates the estimation of EM interactions of graphene-based EEG probes within an MRI environment using the HB approach to accelerate simulations of these microscopic probes. Our simulations show that that the impact of the graphene-based probes on RF transmission and SAR deposition is modest. Further work is needed to optimize computational efficiency, conduct experimental verification using phantoms and evaluate the probe’s electrophysiological performance in the MRI environment.
Suchit KUMAR, Samuel FLAHERTY, Alejnadro LABASTIDA-RAMÍREZ, Anton GUIMERÀ BRUNET, Ben DICKIE, Kostas KOSTARELOS, Rob WYKES, Louis LEMIEUX (London, United Kingdom)
15:40 - 17:10
#47709 - PG419 Comparison of quality control pipeline for skeletal muscle energy metabolism assessed by 31P MRS in patients with muscle weakness at 3T and 7T.
PG419 Comparison of quality control pipeline for skeletal muscle energy metabolism assessed by 31P MRS in patients with muscle weakness at 3T and 7T.
Dynamic phosphorus magnetic resonance spectroscopy (31P MRS) is a noninvasive method for assessment of phosphorus metabolite levels, which reflect mitochondrial respiratory capacity (1,2). However, dynamic data analysis is often hindered by variability in data quality, influenced by patient characteristics and experimental setup. A recently proposed quality control (QC) pipeline developed by Naegel et al. (3, QCS_REF) introduced six key parameters to ensure reliable 31P MRS results in large clinical datasets, such as phosphocreatine (PCr) depletion, the coefficient of determination (R²) for recovery (Rec) and exercise (Ex) kinetic fits of PCr and inorganic phosphate (Pi), the stability of the sum of PCr and Pi during the measurements, exercise time constants (τ) of PCr and Pi, and coefficient of variation (CV) of PCr and Pi at the end of Ex and Rec period. This study tested the applicability and the transferability of this QCS_REF (3,4) to two different research sites equipped with different MR systems (3T and 7T), and different ergometers in patient groups with the focus on frail, elderly and patients with neurodegenerative disease.
The study included six groups in each research center (Table 1): healthy young controls (HCY), elderly healthy controls (HCE) and obese volunteers (OV) at both centers, liver transplant (LT) candidates (LTC), patients 6 months after LT (LT6) and patients with diabetic foot syndrome (DFS) at the site one (S1), and patients with Parkinson’s disease (PD), Alzheimer’s disease (AD) and mild cognitive impairment (MCI) at the site 2 (S2). All subjects provided written informed consent with the participation in the study. The study was conducted in compliance with the principles of the Declaration of Helsinki and with the approval of local ethics committees.
Subjects at both centers were examined on Siemens MR systems (3T @ S1 and 7T @ S2) in a supine position. MR compatible ergometers were used. At 3T, dynamic 31P MRS were obtained by the FID sequence (TR/TE* = 2000/0.4 ms, 420 measurements) and flexible dual 1H/31P surface coil (Rapid Biomedical) fixed under the musculus gastrocnemius. The exercise protocol consisted of a 1-minute rest, a 4-minutes plantar flexion exercise (f = 0.5 Hz) and a 9-minutes of recovery. At 7T, DRESS sequence (TR/TE* = 2000/0.4 ms, 420 measurements) with VOI placed over the gastrocnemius medialis muscle was applied. Dual 31P/1H circular surface coil (Rapid Biomedical) or double-tuned surface coil transceiver array with two 1H channels and three 31P channels was used. The examination protocol included a 2-minutes rest, a 6-minutes plantar flexion exercise (f = 0.5 Hz), and a 6-minutes of recovery. Resistance of the pedal was set to 25-35 % of the individual maximal voluntary force (MVF) at both sites.
31P MR spectra were analyzed using the AMARES fitting routine in the jMRUI v5.0 and time course of 31P metabolites were evaluated at both centers by the same MATLAB script to produce consistent and comparable results of exponential fitting. Metabolic parameters such as mitochondrial capacity (Qmax) and recovery time constants (τREC) of PCr and Pi were calculated. The application of reference quality control limits (QCS_REF) lead to exclusion of substantial part of data for our patient groups and experimental setting. Specifically, only 24 % of all Rec+Ex data and 44 % of Rec period data at 3T (Fig.1) and 32 % of all Rec+Ex and 67 % of Rec data at 7T passed QCS_REF inclusion criteria. Low SNR of Pi signal, reflecting partial T1-saturation at TR of 2s leading unreliable fitting results of Pi dynamics (R2 τPi), was the main reason for the data exclusion. Thus, two new QCS1 and QCS2 were suggested to better reflect our protocol set ups and patient groups. The original criterion of R2 τPi < 0.7 was omitted in QCS1. The sum (R2 τPCr + R2 τPi) < 1.4 was used in QCS2. Using adapted QC, neither τREC nor τEx nor Qmax differed in each patient group (Tab.2). Key parameters recommended in referenced publication (3) proved to be effective for assessing 31P MRS data quality. However, suggested limits need to be modified according to patient groups and TR settings, as shorter TRs used in our experimental protocols reduced Pi SNR, the robustness of its dynamic time course and impacted QC outcomes. Adapted QCS1 and QCS2 thresholds proved to be more adequate in classifying the 31P MRS dynamic data at both 3T and 7T. This project of two research centers successfully evaluated and adapted a joint methodology for the assessment of the quality of dynamic 31P MRS data. It highlights the importance of adjusting QC parameters according to patient characteristics and experimental conditions to ensure reliable and comparable 31P MRS data in different clinical applications.
Dita PAJUELO (Prague, Czech Republic), Radka KLEPOCHOVÁ, Petr ŠEDIVÝ, Monika DEZORTOVÁ, Ivica JUST, Petr KORDAČ, Milan HÁJEK, Martin BURIAN, Pavol SZOMOLÁNYI, Pavel TAIMR, Luděk HORVÁTH, Michal DUBSKÝ, Dominika SOJÁKOVÁ, Martin KRŠŠÁK
15:40 - 17:10
#47695 - PG420 Quantitative susceptibility mapping of the knee cartilage at 7 tesla with aspire multi-echo gradient echo and water-fat total field inversion.
PG420 Quantitative susceptibility mapping of the knee cartilage at 7 tesla with aspire multi-echo gradient echo and water-fat total field inversion.
Knee osteoarthritis is a degenerative disease of the total knee joint in which often the cartilage is affected. Several MRI techniques are available for diagnosing and quantifying the level of osteoarthritis in the knee. Commonly used methods for evaluating the knee cartilage tissue include T2(*) mapping, diffusion-weighted MRI, T1ρ, glycosaminoglycan chemical exchange saturation transfer (gagCEST) and sodium imaging [1]. Collagen composition in knee cartilage is an important feature for the evaluation of osteoarthritis progression or interventions for cartilage repair.
Previous studies have shown that quantitative susceptibility mapping (QSM) is capable of detecting the collagen structural organization in cartilage and canals in juvenile cartilage [2-7]. Although QSM benefits from higher magnetic field strengths, most in-vivo QSM cartilage studies are done at 3 Tesla (3T), and to our knowledge only one study investigated cartilage structure at 7T [2]. In this study, we aim to evaluate QSM of the knee cartilage at 7T, using the ASPIRE multi-echo gradient echo (ME-GRE) sequence [8] and a water-fat total field inversion (wfTFI) QSM processing pipeline [9].
Data sets of five volunteers (aged 26 to 44 years, three males, three left and two right knees), with no known knee damage were acquired on a Siemens 7T Magnetom Plus MR scanner with 1Tx/28Rx channel QED knee coil. After manual B0 shim and B1 power optimization, three series of monopolar 3D ME-GRE with ASPIRE reconstruction were acquired. The three series were acquired twice, once with isotropic voxels and once with higher in-plane resolution. Scan parameters are shown in Table 1. The three acquired phase and magnitude series were combined to one series. A binary mask was created based on the maximum intensity projection across the different echoes of the magnitude data using a threshold of 10% of its maximum intensity [9]. Complex data of the combined series and a fat model of seven fat peaks [10] were used as input in the hierarchical multi-resolution graph-cuts (hmrGC) method [11] to compute water and fat images and a R2* and B0 field map. These processed data were used as input for the wfTFI method of Boehm et al. [9]. A susceptibility map (χ-map) was estimated from the B0 fieldmap first by solving a linear preconditioned TFI problem [12] and second by estimating the χ-map directly from the complex multi-echo data using a water-fat signal model and the initial estimate from the linear TFI. Additional χ-maps were estimated by Projection onto Dipole Fields (PDF) [13] or Laplacian Boundary Value (LBV) [14] background field removal and Streaking Artefact Reduction (STAR-QSM) [15] field inversion as done in previous knee studies [2, 5-7]. The preliminary results are summarized in Figure 1 presenting the χ-maps of one slice of the isotropic acquired data and Figure 2 presenting the χ-maps of one slice of the high in-plane data acquired. Measuring QSM in knee cartilage, using the ASPIRE sequence and performing (wf)TFI show similar results as the PDF + STAR-QSM method. LBV + STAR-QSM show less contrast compared to the other processing methods. Linear TFI and wfTFI look similar. Measuring QSM at 7T in the knee is not trivial, due to increased phase errors caused by the short T2* decay of bone and ligaments, the presence of fat, the pulsation of the Popliteal Artery and due to the small tissue structures of interest. To avoid phase errors caused by different coil sensitivities, an ASPIRE ME-GRE sequence was used [8]. To avoid phase errors caused by the Popliteal Artery, the data was acquired in coronal orientation with phase encoding in right-left and a readout in feet-head direction.
The linear TFI seems to show similar quality as the wfTFI method for the current imaging protocol. One might consider not to perform the wfTFI to save processing time, which will be beneficial for clinical use.
The knee cartilage is thin, ~2 mm, which makes it challenging for QSM processing. The LBV background field removal removes pixels at the edges of the tissue, probably causing signal loss of the cartilage. Therefore, TFI or PDF are preferred.
The results shown are preliminary results of one volunteer. To assess test-retest reproducibility each volunteer was scanned twice (data still undergoing analysis). The lack of an accepted gold standard for susceptibility values makes it difficult to quantify and validate, since different parameters like resolution, echo timing and QSM processing steps yield different χ-maps. In this study we obtained similar results at 7T as reported by Wei at al. [2], using the ASPIRE sequence and (wf)TFI. The evaluated QSM method has potential for knee cartilage assessment with 7T MRI, but it needs further optimization. Validation is needed for clinical practice, as are shorter scan times and faster processing. The added value over the existing methods for knee assessment needs to be investigated in a clinical study.
Esther STEIJVERS-PEETERS (Maastricht, The Netherlands), Laslo VAN ANROOIJ, Marloes PETERS, Dimitrios KARAMPINOS, Jonathan STELTER, Pieter EMANS, Benedikt A POSER
15:40 - 17:10
#47649 - PG421 Detecting CEST Peaks using Curvature Analysis in High Resolution CEST Spectra.
PG421 Detecting CEST Peaks using Curvature Analysis in High Resolution CEST Spectra.
CEST MRI is based on the chemical exchange of labile protons with water protons. One simple approach to assess information of the exchanging protons is by calculating the so-called MTRasym [1]. This can lead to an unwanted superposition with further MT-effects originating in the Z-Spectrum. More precise techniques include Lorentzian and/or polynomial pool-fitting methods [2], which can be limited by low amplitude signal quantification. In this paper, we propose a new method by using curvature analysis, and we evaluate its properties regarding robustness and CEST signal contrast.
The proposed curvature analysis uses a 2-fold numerical derivation of the Z-spectrum in order to detect the offset-specific curvatures of the saturated CEST pools.
This method was investigated in both, Pulseq-CEST simulations [10] and CEST measurements on three human calf muscles within the gastrocnemius. Data was acquired on a 7T Siemens Terra X scanner (Siemens, Healthineers) by using a 24 channel knee coil. For CEST imaging, saturation was achieved by using block-pulses with a 2s saturation time, DC=99 % at three different B1 levels (0.4, 0.6, 0.9 μT). A 3D snapshot readout [11] was used to acquire the offset range from -10 to 10 ppm. B0 and B1 correction were determined by using the field maps from WASABI [9] and applied to the CEST data for post-processing.
Within the Pulseq-CEST simulation, an analysis was conducted on the curvature behavior in response to alterations in both, the concentration of the PCr CEST pool and its exchange rate. The simulated phantom comprised four pools: water, MT, Cr and PCr pool. The simulation parameters for water, MT and Cr were obtained from literature [3-8].
For comparability, all Z-spectra were fitted with a 5-pool lorentzian model. After the derivation, the water peak in the curvature was removed by subtracting the analytical curvature with the fitted parameters of the water pool from the total numerical curvature. As a comparability metric, the relative contrast ratio (RCR) has been calculated according to Eq.1:
RCR_pool=((cur_pool)⁄(cur_H20 ))/((amp_pool)⁄(amp_H20 )) [Eq.1]
Here cur is the curvature and amp is the amplitude of each peak.
In addition to Eq.1, we investigated the theoretical amplification of the noise in the curvature. An example spectrum of the simulation without noise can be seen in Fig.1 with the Z-spectrum (blue) and the corresponding curvature (orange). The curvature spectrum reveals an average RCR_PCr of 9 ± 6 and thus has a higher contrast to water compared to the Z-spectrum. The same spectrum with the addition of noise, smoothing and a five pool fit was simulated in Fig.2. While the curvature gets noisier, the Cr and PCr peaks are also heavily amplified.
We calculated the theoretical increase of noise in the curvature to be
σ_curv=√6/(ω_s^2 ) σ
where σ is the noise in spectrum and ω_s is the sampling rate of the spectrum. Since ω_s is also used during the numerical derivation, this factor cancels out in a SNR and leads to a √6 worse SNR for the curvature, but the error stays proportional to the Z-spectrum error.
Fig.3 shows the measurement within a ROI of the gastrocnemius muscle with the corresponding average Z-Spectrum and curvature for one of the volunteers. The data indicates an RCR_PCr of 8.5 ± 3.9. The curvature contrast demonstrates a relative increase in the water peak of 17%, in comparison to a Lorentzian amplitude contrast of 2%. In addition, Cr (1.9 ppm) and PCr (2.6 ppm) show an amplification compared to the corresponding signals in the Z-spectrum. The removal of the water-peak in the curvature is plotted in red.
In Fig.4, we show the direct comparison of the curvature and Lorentzian maps. The curvature analysis of the Z-spectrum achieves better CEST-water-peak contrast than Lorentzian fitting. While this method amplifies noise by √6, it can detect an RCR_PCr of 8.5 ± 3.9, very close to the simulated result of 9 ± 6. Smoothing the data partially reduces noise.
Curvature analysis enables better CEST peak separation and removes broad peaks like the MT pool, y-axis drift, and other linear influences. It also allows removal of water’s influence, centering the spectrum around zero.
The resulting curvature maps show very homogeneous contrast. Signal increases in the Lorentzian PCr map are also seen in the curvature map. The Cr signal reduction seen in the Lorentzian map is absent in the Cr curvature map, possibly due to a fitting issue, which will be investigated. The relative contrast between the water pool and the PCr pool increase of a factor of 8.5 while only worsening the noise by a factor of √6, together with the very homogeneous maps, makes this method interesting for further analysis. While there are still problems to be solved, like the noise amplification, we believe that this might be a new promising approach for CEST data analysis.
Simon KÖPPEL (Erlangen, Germany), Jan-Rüdiger SCHÜRE, Moritz ZAISS
15:40 - 17:10
#47833 - PG422 Cortical thickness of the human brain with MP2RAGE at 9.4T: methodological challenges and histology.
PG422 Cortical thickness of the human brain with MP2RAGE at 9.4T: methodological challenges and histology.
Cortical thickness is a key biomarker for studying brain alterations, often used to infer neuronal loss or degradation through cortical thinning [1]. However, the lack of a gold standard for in vivo cortical thickness measurements is a challenge for establishing reliable reference values [2]. Methodological innovations, like the availability of ultra-high-field scanners (UHF) or novel processing pipelines further highlight the importance of thorough investigations and comparative studies. Here, we validate the robustness of cortical thickness in UHF-MRI at 9.4T with surface-based and volume-based pipelines and evaluate the correspondence with 3T results and histologically retrieved cortical thickness measures [4, 5].
A total of 39 participants (19 – 74 years, M = 37.5, SD = 17.65), underwent anatomical imaging at a 9.4T whole body MR scanner with MP2RAGE, TI1/TI2 = 900/3500ms; flipangles, FA = 4/6; repetition time TR = 6ms; echo time TE = 2.3ms; volume TR = 9s; and 0.8mm isotropic voxel size. A subset of subjects (N=11; 20–56 years, M = 36, SD = 11.3) also had MP2RAGE scans at 3T with comparable settings [11]. Grey-White Matter Segmentation was performed using FreeSurfer (v.6.0 and v.7.4.0), and the resulting rim of the cortex was used to calculate cortical thickness within FreeSurfer and LAYNII [3] with the LN2LAYERS algorithm. The JueBrain atlas [6] was used for parcellation, available in both volume and in surface space. The cortical thickness obtained in surface-space at 9.4T was compared with histology data using the von Economo parcellation [10]. A vertex-wise paired t-test revealed two significant clusters of cortical thickness differences between 3T and 9.4T, corrected using Monte Carlo Simulation and a cluster threshold of p < 0.001. In the right inferior temporal region, the cortical thickness was higher at 3T (t = 5.48, p = 0.0002), while in the superior frontal cortex, it was lower at 3T (t = −4.06, p = 0.0004). Outside these regions, no significant differences were observed. Comparing volume- and surface-based methods (paired Student’s t-tests), revealed significant differences in mean cortical thickness for the left (t(126) = −51.95, p < 0.0001, Δ = −1.36 mm) and right hemisphere (t(127) = −40.09, p < 0.0001, Δ = −1.34 mm), with a large effect size (d = −4.98; Fig. 1). Finally, a Wilcoxon rank-sum test showed significant differences between MRI-based and histological measurements from v. Economo (W = 327, p < 0.0001) and the BigBrain datasets (W = 209, p < 0.0001) (Fig. 2,3). Correlation analyses confirmed a ~16.1% thinner cortical thickness with MRI. The estimated cortical thickness was generally unaffected by 9.4T UHF, supporting its robustness throughout increasing field strengths, in agreement with [2]. However, there was a significant difference between surface- and volume-based methods, with LAYNII yielding higher thickness values (by 1.34 mm). Since both tools derive cortical thickness from the same cortical rim after GM-WM segmentation, the difference likely results due to methodological differences: FreeSurfer calculates cortical thickness based on the shortest distance between vertex nodes among all neighbouring nodes of the GM and WM surface [7]. LAYNII uses a volume-based approach calculating the thickness as the shortest streamline distance between the outer and inner gray matter borders, which is not necessarily the shortest Euclidean distance [3]. Contrary to prior studies [7–10], MRI-derived cortical thickness significantly differed from histological measurements (von Economo, BigBrain), indicating a systematic effect in in-vivo MRI using MP2RAGE, which cannot be explained by field-strength differences. Figures 3 and 4 support the existence of a systematic effect, as the mean thickness obtained at 9.4T follows a similar region-specific pattern as those determined with histology, with slightly greater discrepancies in regions where the cortex is thinner. Further investigation is needed to identify if systematic differences are due segmentation tools or imaging sequence. Our findings confirm that cortical thickness is a robust measure across field strengths, with only minor differences between 3T and 9.4T, extending prior evidence to ultra-high-field MRI [2]. However, systematically higher cortical thickness found in volume-based in comparison to surface-based methods. These differences highlight the importance of methodological considerations when interpreting cortical thickness measurements across studies. Moreover, comparisons with histological datasets indicated significantly lower cortical thickness values in MRI-derived measurements, contrasting previous studies that reported alignment between in-vivo MRI and histological data [7, 5]. This systematic difference suggests that cortical thickness measurements derived from in-vivo MRI with MP2RAGE is not in line with histological thickness and should be interpreted with caution across literature and calculation pipelines.
Angela OSENBERG (Tübingen, Germany), Jonas BAUSE, Pascal MARTIN, Klaus SCHEFFLER, Gisela E. HAGBERG
15:40 - 17:10
#46651 - PG423 Optimisation strategy for determining refocusing flip angle trains in the multi-echo spin-echo sequence for T2 mapping at 7T.
PG423 Optimisation strategy for determining refocusing flip angle trains in the multi-echo spin-echo sequence for T2 mapping at 7T.
Quantitative MRI is valuable to compare images between scanners and characterise disease progression. In particular, T2 mapping has been proven useful in the detection of myocardial disease [1], pathological tissues in epileptic patients [2], multiple sclerosis [3] and Alzheimer’s disease [4]. T2 measurements are commonly conducted using a multi-echo spin-echo (MESE) sequence typically with an echo train of 180° refocusing flip angles (FA) and a mono-exponential fit of the decaying signal. However, the application of this technique at 7T is limited due to increased B1+ inhomogeneities and specific absorption rate (SAR) levels [5]. To mitigate B1 effects, extended phase graph (EPG)-based fits can be used to model the signal decay [6], [7]. To reduce SAR, trains with low constant refocusing FAs have been used [8]. However, it is not clear which FA train will give the best compromise between T2 precision, image quality and SAR.
The purpose of the present work is to develop a framework to optimise the FA train of the MESE sequence at 7T based on EPG simulations, considering SAR constraints and maximizing the contrast between two tissues. The quality and repeatability of images from the optimised echo train (FAopt) were compared to the images from the 180° echo train.
Framework for FA train optimisation:
We simulated two tissues with a T1 value of 1500 ms and different T2 values corresponding to grey and white matter (60 and 40 ms respectively) [9]. The optimiser was designed to find the combination of FAs (between 0 and 180°), which maximises the area between the two simulated signal decay curves and minimises SAR in the least-squares sense (Figure 1 - Equation 1). The SAR term was defined as the sum of FA squared modulated by a regularisation parameter λ.
Acquisitions:
Four healthy volunteers (3 males, [28-41] y/o) were scanned twice in the same session at 7T (MAGNETOM Terra.X, Siemens Healthineers, Forchheim, Germany) with an 8Tx/32Rx head coil (Nova Medical, Wilmington, USA), taking them out of the scanner between the two runs. The acquisition parameters of the 2D coronal MESE images were: TR=5170ms, echo spacing=9.1ms, 0.8x0.8x2.0mm, 14 slices (50% gap), TA=10:27min. The FAs of the FAopt train are given in Figure 1E. Before each MESE sequence, a 2D B1+ map was acquired using a pre-saturated TurboFLASH (TR=5990ms, TE=1.63ms, FA=5°, 4.0x4.0x5.0mm, TA=13s) [10]. We also acquired a 3D MP2RAGE (T1W) image (TR=6000ms, TE=2.97ms, TI=[802, 2700]ms, FA=[4,5]°,0.6x0.6x0.7mm, TA=7:34min) [11].
Data processing:
B1+ maps were reconstructed [10] and registered to the MESE images using FSL FLIRT [12]. Segmented regions from the T1W images using FreeSurfer [13] were registered to the MESE images using ANTs [14].
A dictionary of EPG-based signal decay curves was generated by varying B1+ (0.2 to 1.3) and T2 (2 to 300 ms) and keeping T1 constant (1500 ms). To compute T2 maps, the B1+ maps were employed to select dictionary curves matching the FA in each voxel. By maximising the cross-product between signals and the dictionary curves, the voxel-wise T2 values were determined.
Statistical analysis:
The fitting error was computed by computing the root mean squared error (RMSE) between data points and fit in each voxel. To compare MESE sequences and runs, the median T2 values were computed in different regions (white matter, deep grey matter, cortex and cerebellum), and linear regressions, Bland-Altman plots and coefficients of variation were used for analysis. The optimisation procedure converges towards a sequence of FAs which minimises the cost function for different values of λ (Fig. 1). Setting λ=0.078 ensures SAR reduction, while mitigating the loss of area between the two curves (T2=60ms and T2=40ms) and gaining slightly in mean signal intensity.
T2 and RMSE maps from the two MESE sequences can be visualised in Fig. 2. SAR is reduced by more than 60% in the FAopt sequence. The visual quality of the T2 maps from the FAopt sequence is not compromised by the reduced FAs and the RMSE is lower overall.
Both types of T2 map have good repeatability, as shown in Fig. 3. However, the MESE sequence with the 180° train has consistently higher T2 values than the MESE with FAopt train (mean difference: 8.90 [6.82,10.98] ms; Fig. 4). T2 maps from the MESE sequence with FAopt train were repeatable and of good quality. However, T2 values were consistently lower than the ones from the MESE with 180° train; the latter also being characterised by a higher RMSE. We noted that the optimised FAs were similar to a train of pseudo-constant FA of 93°, which is lower than previously tested [8]. A flexible EPG-based framework was developed and was used to optimise the FA train of the MESE sequence by maximizing the contrast between two tissues and reducing SAR at 7T.
Emilie SLEIGHT (Lausanne, Switzerland), Ludovica ROMANIN, Gian Franco PIREDDA, Frédéric GROUILLER, Tom HILBERT, Dimitris KARAMPINOS
15:40 - 17:10
#47645 - PG424 T1 mapping with multi-contrast MP-RAGE and 2D GRAPPA at 7T.
PG424 T1 mapping with multi-contrast MP-RAGE and 2D GRAPPA at 7T.
MP2RAGE[1-3] is well-suited to high-contrast T1-weighted (T1-w) imaging of the human brain at 7T due to reduced sensitivity to B1+ inhomogeneity and T2* effects. This is achieved by combining two complex images acquired at different inversion times (TI).
This study expands the quantitative capabilities of MP2RAGE by acquiring additional images at more TIs by extending previous work[4] and employing 2D GRAPPA[5], data sampling in both phase-encoding directions is accelerated, reducing overall scan time and sampling time for each image during spin relaxation after the inversion pulse.
Unlike previous research[6], which used a radial readout, this study utilises a multi-contrast MPRAGE (McMPRAGE) with a Cartesian readout, a straightforward modification of the standard sequence. The Deichmann-Haase correction[7] using SNAPSHOT-FLASH for T1 estimation was extended here to McMPRAGE data. However, it does not account for the incomplete relaxation during the delay time (TD) before each inversion pulse, which is relatively short in typical MPRAGE protocols. To address this, an updated model function, based on more recent work[6], was also explored in a preliminary Monte Carlo simulation.
Scans were performed on a 7T MAGNETOM Terra MRI scanner (Siemens Healthineers AG, Germany) using a custom-built 8Tx64Rx head coil[8] with local ethical approval.
Images were acquired from an agar gel phantom and in vivo with 0.86mm isotropic resolution, TR 4800ms, and GRAPPA acceleration factor 3x2. McMPRAGE was used to scan 4 healthy volunteers (4F, 20-40) with TDs of 2000ms and 8000ms, giving a total acquisition time of 7.9mins and 10.4mins respectively. MP2RAGE images with matched resolution were also acquired (TIs:700ms, 2700ms, TR:5000ms, GRAPPA factor 2) in 8.2mins. Brain, grey matter(GM), and white matter (WM) masks were obtained with FAST[9] in FSL using McMPRAGE's second contrast.
T1 estimation of phantom and in vivo data was performed voxel-wise as a monoexponential, 3-parameter fit using the Deichmann-Haase correction(Figure 1). A 10,000-sample Monte Carlo simulation was performed using MATLAB (R2024b, MathWorks, Natick, MA, USA). In each run, synthetic signal was generated and Gaussian noise was added. The goal was to compare precision and accuracy of T1 estimates from the Deichmann-Haase correction with those derived from an alternative model function that accounts for the incomplete longitudinal relaxation during the TD. The alternative model was tested under 3 fitting conditions: a 2-parameter fit and two 3-parameter fits. Figure 2 illustrates different T1-w contrasts from a single acquisition with TIs (500ms-3000ms) and corresponding T1 maps. Estimated T1 values for TD=2000ms and 8000ms for WM (980±192ms and 1085±232ms) and GM (1562±450ms and 1797±326ms) were comparable for WM but underestimated for GM relative to previously published values (WM 1130±10ms; GM 1940±150ms)[10].
Figure 3 demonstrates that McMPRAGE yields a contrast comparable to the MP2RAGE uniform (UNI) image by using estimated T1 values to generate synthetic images for the TIs used in MP2RAGE.
Figure 4 shows that, in Monte Carlo simulation, Deichmann-Haase correction underestimated T1 while the updated model function yielded a more accurate but less precise estimate with 3-parameter fits, but an accurate and precise estimate using 2-parameter fit. This study demonstrates that extending MP2RAGE to include multiple TIs combined with 2D GRAPPA enables the acquisition of more sample points for T1 fitting without significantly increasing the scan time. Moreover, McMPRAGE can also achieve a contrast similar to MP2RAGE, which is well established in neuroscience studies and clinical practice.
Deichmann-Haase correction used to fit T1 imperfectly accounts for incomplete relaxation during TD. This limitation leads to an underestimation of T1, particularly in tissues with longer relaxation times like GM. While this bias is evident at shorter TDs, at longer TDs, results are comparable with published values and consistent across subjects, as seen in Figure 1.
Monte Carlo simulations further confirm this bias. To address this, parameter fitting using an updated model function was also evaluated for T1 estimation. This function lacked precision for 3-parameter fitting, but when used with a 2-parameter fit, it achieved both accurate and precise T1 estimates.
The superior performance of 2-parameter fitting with the updated model function could be realised in practice by using flip angle values derived from B1+ maps along with inversion efficiency values obtained from Bloch simulations or experimental characterisation as inputs, thereby allowing T1 and ⍴ to be estimated more robustly. This study demonstrates the feasibility of extending MP2RAGE to multiple contrasts with accelerated Cartesian k-space sampling to achieve robust quantitative mapping. Future work will assess the performance of the updated model function when estimating T1 values from McMPRAGE data acquired in vivo.
Janhavi GHOSALKAR (Glasgow, United Kingdom), Graeme A. KEITH, Belinda DING, Natasha FULLERTON, Shajan GUNAMONY, David PORTER
15:40 - 17:10
#47717 - PG425 Optimizing parallel transmission for 7T MRI of the spinal cord with faster acquisitions and higher spatial resolution.
PG425 Optimizing parallel transmission for 7T MRI of the spinal cord with faster acquisitions and higher spatial resolution.
Spinal cord (SC) MRI is challenging due to the small size of the cord. To visualize microstructures [1] and abnormalities [2] in the SC that are undetectable at 3T, both high spatial resolution and SNR are essential, which can be achieved at 7T [3], [4]. However, increasing in-plane resolution can lead to longer acquisition times (TA), or may not be possible due to hardware restrictions. Furthermore, current limitations of 7T MRI include B1⁺ field inhomogeneity and specific absorption rate (SAR) constraints [3], [4]. Those issues can be mitigated using parallel transmit (pTx) [6], recently implemented in the SC [7], [8].
In this work, we use pTx to solve B1+ and SAR issues as well as to improve the trade-off between acquisition time and high spatial resolution in a 3D-GRE sequence, in the context of 7T SC MRI. The proposed method combines signal homogenization within a region of interest (ROI) with signal suppression outside the ROI, which allows a reduced field-of-view (rFOV) without aliasing artifacts [6]. The rFOV technique was implemented in a phantom and was evaluated for reduced scan time or increased spatial resolution.
All experiments were conducted on a 7T Magnetom TERRA (Siemens Healthcare) using an 8 Tx/Rx cervical coil (Rapid Biomedical) loaded with a head and neck phantom (SPEAG, Zurich, Switzerland).
The gradient ascent pulse engineering (GRAPE) algorithm [9] was used to optimize pTx pulses [7] in the rFOV mode. B1⁺ maps (TurboFLASH satTFL) [7], [10] and B0 maps (3-echo GRE) were used as inputs for the optimization. The optimization was performed using a magnitude least-squares approach with: 500 µs pulse duration, 1000 iterations, 10° target flip angle, 10 µs time step and a slew rate constraint of 195 T/m/s.
The optimized RF pulse was used in a 3D-GRE sequence with an initial spatial resolution of 0.78×0.78×2.5 mm³ and a FOV of 184×200×240 mm³, which were chosen to limit TA to ~10 min. Sequence parameters are provided in Table 1. The rFOV approach was used to increase resolution or reduce TA by decreasing FOV to 80×80×200 mm³.
The signal-to-noise ratio normalized to acquisition time (SNRN) was used to compare the two rFOV configurations. In addition, flip angle normalized root mean square error (FA NRMSE) was evaluated to assess the accuracy of the excitation profile. RF pulse optimization using the GRAPE method enabled effective selective excitation in rFOV mode. The signal was largely suppressed outside the (ROI), and maintained homogeneous within the ROI, as shown by the simulated magnetization maps (Fig. 1A).
A quantitative analysis showed a normalized root mean squared error (NRMSE) of 21% for the flip angle (FA) within the ROI. Outside the ROI, the FA cancellation NRMSE was ~8%, demonstrating effective signal suppression. All optimized pulses complied with the imposed physical constraints, particularly regarding the specific absorption rate (SAR) (Fig. 1B), and were calculated in less than 15 minutes.
In Fig.2A (circularly polarized (CP) mode), the baseline acquisition yielded a SNR of 49.57 and a SNRN of 2.02. When improving spatial resolution using rFOV (Fig. 2C), the SNR dropped to 23.84 (−52%) and SNRN to 1.00 (−51%) compared to the CP mode. However, this high in-plane resolution (0.2 × 0.2 mm²) was achieved within a similar TA (~9.5 min), which would not be feasible with a full-FOV excitation. When applying the rFOV strategy to reduce acquisition time to less than 4 min (Fig. 2D), the SNR decreased to 30.62 (−38%), but SNRN remained comparable (2.04 vs. 2.02).Overall, rFOV enabled either substantial TA reduction or resolution increase with controlled SNR loss, while maintaining clinically acceptable scan durations. In this study, we successfully implemented a rFOV excitation using pTx with a SC radiofrequency coil on phantom at 7T. This approach allowed either a 2.7-fold reduction in TA, or an increase in in-plane resolution up to 0.2 × 0.2 mm² within a clinically acceptable TA (~9 min), without introducing aliasing artifacts.
With full-FOV excitation 3D-GRE and the sequence parameters used in this study, the maximum spatial resolution was only 0.66 × 0.66 mm2 due to scanner limitations. The rFOV pTx method thus enables efficient use of the available SNR and improves imaging flexibility depending on clinical priorities (resolution vs. speed).
However, a decrease in SNR and SNRN was observed at high-resolution. A precise evaluation of SNR requirements will be necessary for in vivo application to ensure sufficient image quality. Residual signal outside the ROI was also observed, likely due to B1⁺/B0 map inaccuracies. Combining rFOV excitation with coil sensitivity encoding could improve robustness to imperfect cancellation [9].
Future work will focus on assessing in vivo SNR requirements to guide the trade-off between spatial resolution and acquisition time, as well as evaluating rFOV applications in healthy subjects.
Charles BETEMPS (Marseille), Vincent GRAS, Joseph BREGEAT, Virginie CALLOT, Aurélien DESTRUEL
15:40 - 17:10
#46665 - PG426 Dual-venc phase contrast mri using fast interleaved radial mixing (pc-firm).
PG426 Dual-venc phase contrast mri using fast interleaved radial mixing (pc-firm).
Phase contrast (PC)-MRI has been widely used to detect and characterize blood flow abnormalities either for neuro- or cardiovascular diseases. However, conventional PC-MRI is restricted to the specific flow sensitivity range of the selected velocity encoding (VENC). Consequently, regions with high velocity differences, e.g. neurovascular vessels or aortic branches, poses inaccuracies in the estimated flow velocity. To overcome this limitation, dual-VENC approaches acquiring two sets of flow maps with low and high VENC have been introduced [1,2]. The prolonged acquisition time of the dual-VENC can be reduced using parallel imaging [3]. However, most approaches use conventional Cartesian encoding for image acquisition. In this work, we present a novel approach to accelerate dual-VENC PC-MRI using radial spatial encoding and k-space weighted image contrast (KWIC) [4]. The introduced phase contrast fast interleaved radial mixing (PC-FIRM) approach is based on an intelligent combination of radial projections with different flow sensitivities to create dual-VENC flow velocity maps.
The prototype PC-FIRM sequence is based on a spoiled radial FLASH sequence with flow sensitive bipolar gradients. Schematic illustration of the proposed sequence design and k-space mixing scheme is shown in Figure 1. Based on the KWIC idea, the PC-FIRM sequence acquires radially encoded k-space lines with interleaved polarity (positive, negative) and strength (low VENC, high VENC) of flow sensitive gradients. In theory, the k-space data of each velocity encoding can be reconstructed separately to create conventional PC-MRI. In PC-FIRM, artificial k-spaces containing low frequency spatial information only from the desired VENC and high frequency spatial information from all acquired data are combined and reconstructed using a conventional gridding algorithm. The proposed sequence was implemented on a 0.57 T MRI system (Magspec, Pure Devices, Germany) with 10 mm bore size and maximum gradient strength of 1.5 T/m. To validate the obtained flow velocities of PC-MRI, an experimental setup (Figure 2) consisting of two tubes with different inner diameters (1 mm and 2 mm), filled with a solution of water and gadolinium based contrast agent (Vasovist, Bayer Schering, Germany) is used. With a syringe pump (KL-702, KellyMed, China) different flow velocities can be simulated in the tubes. The adjusted velocity in the big tubes is 0.13 cm/s, whereas the velocity in the small tubes is 3.18 cm/s. PC-FIRM was obtained with following acquisition parameters: TR/TE = 30/8 ms, flip angle = 43°, field of view = 10 × 10 mm², spatial resolution = 0.156 × 0.156 mm², slice thickness = 5 mm, low VENC of 0.25 cm/s and high VENC of 5 cm/s. In addition to flow velocity maps calculated form PC-FIRM, reference maps were acquired using flow sensitive radial FLASH. Figure 3 shows the estimated flow profiles obtained using the PC-FIRM approach for a low VENC of 0.25 cm/s (left) and high VENC of 5 cm/s (right). As expected, phase wrapping is observed when the VENC is set too low, e.g. in the case of the small tube at low VENC. Nevertheless, the characteristic parabolic shape of laminar flow can be observed in all acquisition schemes and tubes. For quantitative analysis of the big tubes, the acquisition with low VENC is considered, whereas the small tubes (characterized by high flow velocities) are evaluated using high VENC. The mean flow velocity of the big tubes was 0.12 cm/s with both methods. In the small tubes, the reference method yielded a mean velocity of 2.82 cm/s, compared to 2.68 cm/s obtained with PC-FIRM. The proposed PC-FIRM is a fast and robust alternative to Cartesian dual-VENC sequences, incorporating the advantageous of radial encoding to facilitate the acquisition time of PC-MRI. In contrast to other acceleration techniques, PC-FIRM weights coequal k-space data of different VENC to fulfill the Nyquist criterion and fasten the image acquisition. Compared to conventional single-VENC acquisition, dual-VENC could be established in one fourth of the acquisition time. One limitation arises from the characteristics of the selected experimental design, leading to laminar flow profiles with high frequency components. These high frequency components can lead to underestimated flow profiles especially in small tubes and high flow rates. Further acceleration can be achieved by combining PC-FIRM with parallel imaging. Unfortunately, the used MRI system is limited to a single-channel receiver coil. In principle, PC-FIRM can be extrapolated to a multi-VENC approach by combining more than two VENCs without prolonging the scan time. The introduced dual-VENC phase contrast MRI will increase the accuracy and reduce the acquisition time of flow maps in regions with high velocity differences like neurovascular vessels and aortic branches. Since, some methodological improvements are still necessary, the potential of the PC-FIRM approach could be shown in this feasibility study.
Maurice RÜGER (Lüdenscheid, Germany), Tobias KRAATZ, Jens GRÖBNER, Amir MOUSSAVI
15:40 - 17:10
#46167 - PG427 Would a ROI-specialized SCOTCH Multi-Coil Array improve B0 shimming of the temporal lobes?
PG427 Would a ROI-specialized SCOTCH Multi-Coil Array improve B0 shimming of the temporal lobes?
Ultra-high field functional Magnetic Resonance Imaging (fMRI) with EPI suffers from significant B0 field inhomogeneities. The Multi-Coil SCOTCH Shim Array was designed to reduce these inhomogeneities in the whole brain (WB)[1], based on a PCA of optimized stream functions (OSF). Moreover, based on OSF targeting specific regions of interest (ROIs), a previous simulation study has hinted that ROI-dedicated hardware could improve shimming performance in such ROIs compared to a WB-dedicated device [2]. While some local MCAs have targeted ROIs such as the frontal lobe [3,4], no existing device specifically addresses the temporal lobes (TL).
In this work, based on the SCOTCH methodology [1,5], we design an MCA for shimming TLs while controlling B0 field excursions elsewhere, in particular to prevent unexpected image aliasing in case of partial brain imaging. We name it TL-SCOTCH and compare its performance to a WB-SCOTCH when focusing shimming on the TL.
We used a database of 96 B0 fieldmaps obtained at 3T, scaled to simulate 11.7T conditions (mean std after scaling: 115.0 Hz). Subject-specific masks for the temporal lobes were created using linear registration of the Destrieux Atlas’ TL, followed by dilation to ensure coverage despite registration inaccuracy.
Consider a Multi-Coil Array of Nc coils placed around a subject's head. The shimming problem aims to minimize B0 inhomogeneities by finding the currents I that minimize the variance (σ²) of the corrected magnetic field: Iopt = arg min σ²(b + CI), with a constraint on individual current intensity.
Here, b is the original Nv-vector fieldmap, and C is the Nv x Nc matrix relating the current inputs to the magnetic fields produced by each coil.
The design problem consists of designing a set of coils that have the best shimming capabilities. In our study, we focused on MCAs based on SCOTCH design methodology, which relies on Singular Value Decomposition of stream functions computed through an optimization problem similar to the shimming problem, taking total power as a regularization term [5].
We modified the loss function using a weighted variance to target a specific ROI: voxels inside the ROI were weighted 1, while voxels outside were weighted by a parameter w ∈ [0;1]. Thus, w = 1 gives a whole-brain (WB) SCOTCH design, while decreasing w from 1 to 0 progressively increases focus on the ROI.
First, we assessed the shimming performance of a WB-SCOTCH for 30 w values ranging from 1 to 1e-3. Correction performance was computed as the relative B0 inhomogeneity reduction (in %): η = 100 x (1 - σ(b0 + bcoils)/ σ(b0)).
From these simulations, we identified an optimal w-value (maximal performance in ROI with no decrease in homogeneity outside) and designed a TL-SCOTCH by solving the design problem with this w value. The performance of this new design was compared to that of WB-SCOTCH.
Both the TL- and WB-SCOTCH designs consisted of 48 coils, each represented by a 20-turn winding placed on one of three cylindrical formers (radii: 14.10, 14.95, and 15.80 cm; length: 30 cm) (see [1]). Optimal currents were constrained by a current limit of 5A. The performance of the WB-SCOTCH in the temporal lobes increased from 18.9% with whole-brain shimming (w = 1) to 36.4% with a targeted approach (w = 0.01), reaching a plateau beyond this value (Fig.1). Despite a more homogeneous field inside the ROI, large areas of high inhomogeneity remained (Fig.2). The subsequent TL-SCOTCH was designed with the identified w-value of 0.01. This design consisted of 48 coils presented in Fig.3 and achieved only a modest improvement in focused TL shimming performance (36.8%) compared to WB-SCOTCH (36.4%) (Fig.4). For both designs, shimming performance with w = 0 showed very small improvement compared to w = 0.01 but resulted in significant decrease of inhomogeneity outside the temporal lobes. The observed plateau in B0 shimming performance (Fig.1) indicates a limit in achievable performance. This could be due to hardware limitations such as current or spatial constraints (cylindrical former and distance from the subject’s head), or to a fundamental limit in B0 correction, as indicated in a previous study [2]. The modest improvement in focused-shimming performance with the TL-SCOTCH design compared to WB-SCOTCH suggests that the flexibility brought by the multiplicity of the coils is sufficient for WB-SCOTCH to produce a wide range of field patterns.
For both designs (TL and WB), choosing w = 0.01 allows reaching optimal performance without degrading homogeneity in the rest of the brain, preventing aliasing artifacts.
Further work, not presented in this abstract, shows similar results on SCOTCH MCA designs for ventral temporal cortex and prefrontal cortex. Our results suggest that targeted shimming with a whole-brain SCOTCH can significantly enhance B0 correction in specific ROIs, such as the temporal lobes, while controlling for inhomogeneity in the rest of the brain, without the need for a ROI-specific SCOTCH.
Ulysse BOUREAU (Paris), Qi ZHU, Alexis AMADON
15:40 - 17:10
#47794 - PG428 Design of Controlled Polyphasic Plastisol Phantom: Magnetic Resonance Elastography Calibration and Reproducibility.
PG428 Design of Controlled Polyphasic Plastisol Phantom: Magnetic Resonance Elastography Calibration and Reproducibility.
Phantoms are essential in medical imaging, particularly for calibrating measurement systems. While tissue-mimicking models are commonly used for medical training and surgical procedure enhancement, magnetic resonance elastography (MRE) still lacks phantoms that combine diverse magnetic and mechanical properties. This limitation impedes the analysis of complex samples where precise geometric and anatomical structures are not known in advance. In this work, we develop polyphasic phantoms with controlled properties, suitable for characterizing heterogeneity and studying interfacial effects.
We generated mechanical waves (700Hz) using a compression driver directed toward a cylindrical sample (30mm diameter, 17mm height) placed on a horizontal xz-plate [1]. Horizontal oscillations, perpendicular to the cylinder axis, induced vertical shear waves along y-axis throughout the sample, see Figure 1.
The acquisition parameters are: B0 = 9.4T (MRI system Agilent), 8 delay offsets, 3 spatial directions, 4 Gauss/cm bipolar sinusoidal gradient. Fast spin echo (4 ETL), TR/TE=3000/22ms, 18 slices (0.7mm thickness, no gap), 2 averages, 64x64 matrix, 45x45mm FOV, 0.7mm isotropic resolution.
The shear stiffness was determined using both inversion NLI [2] and AIDE [3] with two configurations: a fine 5×1×5 grid with random shifts for node averages, and a coarser 1×1×1 grid assuming element homogeneity.
To evaluate MRE reconstruction quality, we developed four calibrated polyphasic phantoms of increasing complexity: sectorial designs from simple bisection (P2) to eight-sector division (P8), plus a checkerboard variant (C3) with a regular 3×3 grid pattern (Figure 2-Magnitude).
All polyphasic structures were fabricated by cutting homogeneous plastisol samples with different softener ratios (r) using custom 3D-printed molds for precise alignment. Components were reassembled through controlled thermal welding, exploiting plastisol's thermoplastic properties to create integrated solid blocks (Figure 2). Increasing the softener content from sample #1 to #9 affects the 1H NMR relaxation as follows: T1 remains relatively constant, varying slightly from 553 to 515 ms, whereas T2 increases linearly with r, ranging from 19.5 to 26.1 ms (data not shown). The quasi-linear r-T2 correlation suggests T2 could serve as an indirect mechanical property marker. Shear stiffness decreases with increasing r, following a slightly non-linear trend (33-10 kPa, Figure 3). Both methods yield consistent shear modulus values (μ±∆μ) with good agreement across samples. AIDE-fine results average 3.7 kPa higher than NLI, while NLI shows superior reproducibility (mean deviation: 0.5 kPa vs. 1.3/1.6 kPa for AIDE).
Contrast analysis between compositions with varying softener ratios in samples #(1-7, 3-5, 6-7) using Cohen’s formula reveals that shear stiffness exhibits at least 46% higher contrast than magnetic properties (T1, T2) and damping ratio across all tested regions (Figure 4).
Comparison of stiffness values (μ±∆μ) between homogeneous and polyphasic phantoms (Figure 2) reveals excellent agreement for the P2 phantom (NLI: 30.3±0.7 kPa vs. 29.7±1.1 kPa at #1 and 11.4±0.4 kPa vs.11.5±0.8 kPa at #8; AIDE: 36.5±2.9 kPa vs. 31.8 kPa and 13.6±2.7 kPa vs. 14.5 kPa respectively), while the more complex P8 phantom shows good overall correspondence with a slight underestimation of stiffer regions and overestimation of softer ones (NLI: 24.3±1.8 kPa vs. 27.5±0.9 kPa at #2 and 16.6±1.8 kPa vs. 15.5±0.4 kPa at #6; AIDE:26.8±3.0 kPa vs. 30.3 kPa and 19.0±2.6 kPa vs. 18.6 kPa respectively). Welding process evaluation comparing samples with identical r ratios, with and without welding, showed nearly identical results (9.9±0.5 vs. 10.1±0.5 kPa for NLI), demonstrating that the process doesn't significantly degrade sample properties (data not shown). In polyphasic samples, both algorithms show systematic tendencies, underestimating μ in stiffer regions while overestimating in softer ones with higher standard deviations. For instance, C3 phantom contrast #3-5 regions dropped significantly from 8.83 in homogeneous samples to just 1.53. Both algorithms agree well in homogeneous samples with consistent shape reconstructions, but NLI excels in polyphasic samples with more accurate boundaries. As sample complexity increases, contrast between regions diminishes, yet both algorithms still distinguish different zones effectively despite shifted absolute values. For tissue characterization, relative stiffness values remain adequate for comparison despite reduced contrast. Plastisol's reversible thermal properties withstand multiple heating-cooling cycles without degradation, enabling complex phantom construction. Its viscoelastic properties can be tuned by adjusting softener ratio (r) for precise tissue-mimicking. Mechanical contrast (μ) significantly outperforms conventional imaging contrasts (M0, T1, T2), making plastisol phantoms excellent candidates for refining MRE experiments.
El-Farid OUSSEN (Montpellier), Christophe GOZE-BAC, Maida CARDOSO, Sebastien ROUSSET, Yvan DUHAMEL, Gille CAMP, Eric ALIBERT, Alain CHARBIT, Jonathan BARES, Elijah VAN HOUTEN, Marion TARDIEU, Bertrand WATTRISSE
15:40 - 17:10
#47929 - PG429 Evaluation of the Safety of an RF Array for Human Head MRI at 9.4T in the Presence of an EEG Cap.
PG429 Evaluation of the Safety of an RF Array for Human Head MRI at 9.4T in the Presence of an EEG Cap.
Ultrahigh Field (UHF) MRI (B0≥7T) allows for increased image resolution and SNR in brain studies compared to clinical systems with lower fields. One of the most popular methods of MRI brain functionality study, fMRI, has low temporal resolution. In contrast, electroencephalography (EEG) offers higher temporal resolution and enables tracking and comparing neural processes during an MRI study[1]. However, since the UHF wavelength becomes comparably lower than 1 m in free space and 10 cm in brain tissues, the presence of a conductive EEG cap can increase the local specific absorption rate(SAR). Therefore, it is essential to carefully evaluate RF safety risks of combined MRI and EEG studies at UHF[2].
In this work, we performed numerical simulations of a setup combining a 16-channel Tx-only loop array and a 31-channel EEG-cap (Fig. 1A, B) using CST Studio 2024 (Dassault Syst.). Simulations were performed using the Duke voxel model, all studies were performed in CP mode. To evaluate the results of numerical simulations, we performed B₁-field measurement (AFI[4]) on a home-made gel phantom (Fig. 1C) in the presence of a 31-channel EEG cap (Easycap, Brain Products) with copper leads, to estimate their influence on the performance of the Tx-array at 9.4 T. The advantage of this phantom is its conductive surface, which is able to mimic human skin impedance, allowing realistic EEG operation. The phantom is composed of sucrose, gelatin, salt, and water. It has a relative permittivity of 36.8 and a conductivity of 0.59 S/m. All measurements were performed using a Siemens Magnetom+ 9.4T full-body MR scanner. Numerical simulations with the phantom and the EEG setup showed a 14% decrease in B₁⁺ (Fig. 2A, C), averaged over a 130 mm region of the voxel model down from the top (corresponding to the human brain size). A similar B1+ drop was observed in the experiment. Fig. 2 B and D, Fig. 3A and C show SAR10g and B1+ maps in the sagittal plane through SAR10g maximum, numerically obtained without the EEG cap for the Duke voxel model. pSAR10g locations are marked with black arrows. Fig. 3 B and D present the results with the EEG cap. In the presence of the cap, the peak SAR10g value in voxel models moved under one of the electrodes and increased from 0.39 to 0.43 W/kg. The RF electromagnetic field, created by coils, excites currents on the surface of the EEG leads and creates a secondary field. This disrupts B₁⁺ under the cap in the superior region of the phantom and voxel model, decreasing the quality of the MR image.
To address this limitation in multimodal imaging, EEG leads must be shielded from the RF coil to prevent coupling. For example, one can wrap the leads with a metal shield incorporating an RF trap to create high impedance for EM wave propagation along the leads, introduce resistors or RF chokes, or replace copper leads with a material with high resistance[3].
The local increase in SAR10g in the voxel model can be explained by numerical overlap between conductive electrodes and voxels. The solver assigns RF electrical current from the electrode directly to the voxel of tissue, without accounting for resistance between the electrode and the tissue. In the future, this issue will be solved by adjusting simulation parameters. The introduction of an EEG cap during MRI leads to a decrease in image quality due to B₁⁺-field drop. It also causes a moderate increase in pSAR10g at 9.4 T. Under these conditions, health risks can be mitigated by applying standard SAR safety control protocols.
Egor BEREZKO (Tuebingen, Germany), Sebastian MUELLER, Joern ENGELMANN, Vinod KUMAR, Georgiy SOLOMAKHA, Jonas BAUSE, Nikolai AVDIEVITCH, Klaus SCHEFFLER
15:40 - 17:10
#47868 - PG430 Feasibility at 7 Tesla of spin- and gradient-echo dynamic susceptibility contrast MRI for cerebral microvascular characterization.
PG430 Feasibility at 7 Tesla of spin- and gradient-echo dynamic susceptibility contrast MRI for cerebral microvascular characterization.
Dynamic Susceptibility Contrast (DSC) MRI is a valuable tool in the diagnosis and follow-up of patients with adult-type diffuse glioma [1]. While the recommended approach for DSC-MRI in clinical practice is to use a gradient-echo sequence, combined spin- and gradient-echo (SAGE) techniques [2–4] could enhance the characterization of such tumors. Despite the clinical feasibility of SAGE-DSC at 3T and the anticipated superior DSC performance at higher field strengths [5], there are no studies that have explored the feasibility of SAGE-DSC at 7T in humans. This study aims to address this gap by developing an optimized protocol for SAGE-DSC at 7T and investigating its feasibility for cerebral microvascular characterization in patients with glioma.
This prospective study was performed as part of the IRB-approved Vascular Signature Mapping (VSM) study (ClinicalTrials.gov ID: NCT05274919). All patients provided written informed consent. Eight patients under surveillance after glioma treatment (4 female, mean age 56 years) were scanned on a 3T MRI-system (Philips Achieva or GE Signa Premier) and on a 7T Philips Achieva MRI-system. At 7T, the contrast agent protocol and the SAGE-DSC scan parameters varied across patients for protocol optimization. rCBV maps at 3T and 7T were obtained with IntelliSpace (Philips Healthcare) applying BSW leakage correction [6]. The resulting maps were visually compared to each other and to the radiologist’s interpretation of the clinical data (ASL, T1w, T2w). Furthermore, 3T and 7T data were compared using ΔR2(*) hysteresis loops [2]. Lastly, an open-source simulation model [8] was used to investigate the behavior of the spin- and gradient-echo signals for varying vessel radii [9] at higher field strength. Protocol optimization: Starting from the clinical protocol at 3T, the contrast agent dose had to be reduced from 15 mL to 10 mL 0.5M Clariscan to achieve a time course with a sufficiently sharp bolus passage. Contrast administration was performed through manual injection (approx. 3 mL/s) as no 7T compatible power injector was available. The sequence parameters were set to TE(GRE1/GRE2/SE) = 10.7/51.2/81.6 ms, 11 slices and 90 time points.
Perfusion assessment: Two patients could not be assessed due to insufficient data quality of the 7T data. In one patient, the clinical data showed no increased perfusion while this was visible on SAGE-DSC, and later follow-ups showed indications of tumor progression at this same location. The radiologist’s observations based on the clinical data agreed with the SAGE-DSC data in the rest of the patients. One of these patients, shown in Figure 1, exhibited elevated perfusion, which was better discernible on 7T.
Vascular architectural imaging: In patients 6-8, protocol parameters and data quality were sufficiently consistent to allow for characterization of the microvasculature. The hysteresis loops (Figure 2) show a comparable shape and direction at 3T and 7T.
Signal simulations: From the signal simulations, ΔR2(*) was computed for different vessel radii at 3T and 7T. The results are shown in Figure 3. The specificity of the spin-echo to the microvasculature – indicated by the peak in ΔR2 - appears to shift towards smaller radii with increasing field strength. Additionally, the gradient-echo seems to become more specific to smaller vessels at a higher field strengths, as shown by the peak in ΔR2*. The first aim of this work was to establish an optimized protocol for SAGE-DSC at 7T in terms of contrast agent administration and imaging parameters. We have observed that good data quality could be achieved with a dose reduced to 2/3rd of the dose at 3T. Furthermore, a trade-off between temporal resolution and coverage was found for the obtained parameter settings. The total scan time could be decreased by lowering the number of time points to 60, but this could affect the performance of the BSW leakage correction [6].
The second objective of this work was to evaluate the feasibility of vascular characterization at 7T using SAGE-DSC. Overall, the rCBV maps generated at 3T and 7T showed good agreement with the clinical evaluation, and the hysteresis loops showed similar behavior at 3T and 7T. Strikingly, the signal simulation showed the onset of microvascular specificity in the gradient-echo signal at higher field strengths. It is unclear how this can be explained by the theory of diffusion narrowing regime [3] and further investigation is warranted.
A limitation of this preliminary study is the small number of subjects and further validation is necessary. This study optimized a scan protocol for SAGE-DSC at 7T. In addition, promising first results considering its application for microvascular characterization were obtained. Indeed, the rCBV maps and hysteresis loops showed similar behavior at 3T and 7T. Further validation in a larger group of preoperative patients with glioma is necessary.
Karen VAN DER WERFF (Rotterdam, The Netherlands), Danielle VAN DORTH, Krishnapriya VENUGOPAL, Esther WARNERT, Dirk POOT, Frans VOS, Marion SMITS, Chad QUARLES, Juan HERNANDEZ-TAMAMES, Johan KOEKKOEK, Jeroen DE BRESSER, Matthias VAN OSCH
15:40 - 17:10
#47690 - PG431 Quantitative susceptibility mapping of the cervical spinal cord at 7 Tesla.
PG431 Quantitative susceptibility mapping of the cervical spinal cord at 7 Tesla.
Quantitative susceptibility mapping (QSM) is a non-invasive method of quantitatively mapping magnetic susceptibility in vivo. Ultra-high field MRI enhances the susceptibility contrast and increases signal-to-noise ratio (SNR) of the MR signal [1,2]. QSM of the brain at ultra-high field is a promising technique to detect iron accumulations, associated with several neurodegenerative diseases [1]. Iron accumulation in the spinal cord is also an indicator of neurological diseases, such as multiple sclerosis (MS) [3]. However, no studies have yet explored QSM of the spinal cord. MRI of the spinal cord is more challenging than the brain, due to small cross-sectional area and strong artifacts from static B0 inhomogeneities and physiological B0 fluctuations. Methods developed for the brain may not be optimal in the spinal cord. This work aimed to explore and optimize parameters of existing QSM acquisition sequences and algorithms for application to QSM of the spinal cord at 7 Tesla.
Data was acquired in 5 subjects on a 7T Magnetom Terra system (Siemens Healthineers), using a 1Tx, 24Rx C-spine coil (MRI.TOOLs). A 3D multi-echo GRE sequence (0.75 mm isotropic resolution, TR 27ms, FOV 144mm, R = 2, FA 17°, ASPIRE coil combination [4]) optimized for brain QSM was used as starting point. In one subject, the sequence was acquired repeatedly with one parameter changed per acquisition (Fat Suppression (FS), Phase Stabilization (PS), receive bandwidth 260 vs 300 Hz/pixel, signal scaling factors, Partial Fourier (PF)) to optimize the acquisition. Finally, the optimized sequence (TE 4.08/9.18/14.28/19.38, no FS, PS, 260 Hz/pixel, signal scaling, no PF) was acquired with 8 repetitions in 4 subjects.
Spinal cord masks were segmented using sct_deepseg in Spinal Cord Toolbox [5]. A gray matter (GM) mask was obtained with sct_deepseg_gm, and a white matter (WM) mask was calculated by subtracting the GM mask from the spinal cord mask. The SNR of each acquisition was calculated within the WM mask on successive volumes of eight slices.
Different algorithms for calculating QSM images were evaluated using the SEPIA toolbox in MATLAB R2024b. All phase images were unwrapped using SEGUE. Next, all parameters in five background field removal algorithms (LBV, PDF, SHARP, RESHARP, VSHARP) were tested and compared using visual inspection, standard deviation, and contrast-to-noise (CNR). Using the local field from the PDF background field removal, four dipole inversion algorithms (iLSQR, FANSI, TKD, MEDI) were compared using visual inspection, standard deviation, and CNR. Standard deviation and CNR were calculated on eight-slice volumes before averaging over the whole volume.
Averaging over repetitions was performed after the QSM pipeline. With the optimized QSM pipeline (SEGUE, PDF, iLSQR), the standard deviation and CNR in the resulting QSM images were evaluated to assess the effect of averages. Magnitude and phase images acquired with different imaging parameters showed comparable visual image quality. However, the SNR of the magnitude images without FS, with 260Hz bandwidth, and no PF had consistently higher SNR (Fig. 1).
Fig. 2 shows results from different algorithms in the QSM pipeline. SEGUE effectively removed all phase wraps, without affecting phase values in areas without wraps. Background field removal with PDF yielded lower standard deviation and higher CNR in the local field compared to the other algorithms. Of the four dipole inversion algorithms tested, iLSQR performed best both in terms of standard deviation and CNR. The results of the algorithm comparison were consistent between subjects, despite large inter-subject CNR variability.
Fig. 3 shows standard deviation and CNR of the QSM images with different numbers of averages. As expected, more averages decreased the standard deviation. The CNR generally increased with more averages, but with a tendency to plateau in individual subjects. CNR values were clearly higher in two of the subjects (0.86, 0.45, 1.05, 0.05). The QSM images from the optimized pipeline are shown in Fig. 4. Visually, contrast between gray and white matter was clearly distinguishable in two of the subjects, corresponding to the subjects with higher quantitative CNR values. The aim of this work was to optimize existing QSM algorithms for use on cervical spinal cord data. Background field removal with PDF and dipole inversion with iLSQR performed consistently better than other algorithms. With eight averages, the resulting QSM images showed clear GM/WM contrast in half of the subjects. The results demonstrate the potential of QSM as a promising technique, while highlighting challenges in the spinal cord. Further work to reduce physiological noise may improve the quality of the data, while targeted algorithms to remove susceptibility effects from vertebrae may improve background field removal.
Karen Johanne ØFSTAAS (Trondheim, Norway), S. Johanna VANNESJO
15:40 - 17:10
#46568 - PG432 Quantification of the arterial input function during PCASL perfusion imaging using ASLIF – First results.
PG432 Quantification of the arterial input function during PCASL perfusion imaging using ASLIF – First results.
Pseudo-continuous arterial spin labeling (PCASL) is commonly used in in ASL experiments due to its superior signal-to-noise ratio compared to other ASL sequences like pulsed labeling techniques. However, the PCASL labeling process is prone to several influencing factors like local field inhomogeneity within the labeling area or blood flow velocity. Therefore, apparently underperfused vascular regions measured with PCASL might not necessarily reflect an actual perfusion deficit. The arterial spin labeled input function method (ASLIF) allows to qualitatively visualize the PCASL labeled bolus during the labeling process itself without the need for separate measurements, restrictions to the labeling scheme or alteration to the blood magnetization, in contrast to existing methods [1-3]. In this work, first results of an ASLIF quantification model are presented which enables quantitative labeling efficiency (LE), flow velocity and T1 estimations.
Model:
Equation (a) in Fig. 2 shows the established signal equation of the acquired ASLIF signal with ∆S denoting the control-label subtracted ASLIF signal, β a proportionality constant, M0b the equilibrium magnetization of the blood/fluid, LEeff the effective PCASL labeling efficiency, Vavg the average flow velocity, c(t) being an averaged delivery function across the velocity distribution, T1b the longitudinal relaxation time of the blood/fluid and TT(v) the transit time between PCASL and ASLIF slice. The model can be seen as a modification and extension to a previously published model for ASL signal in large arterial vessels [4].
To eliminate the pre-factor, a short reference measurement is carried out at the beginning of the scan in which the PCASL labeling is replaced by a PASL inversion, as this has a more robust labeling efficiency.
By combining both signal models a model fit for LEeff, Vavg and T1b can be established as depicted in Equation (b) of Fig. 2 with ∆Sref and LEref being the ASLIF signal and labeling efficiency of the reference measurement.
Phantom imaging:
The flow phantom QASPER [5] (Fig. 1) was examined with an in-house developed ASLIF sequence [4] on a 3T MR scanner (MAGNETOM Skyra, SIEMENS Healthineers, Erlangen, Germany). The ASLIF images, as depicted in Fig. 1, were acquired with the following parameters: 4 Hadamard encoding cylces [6,7], subbolus duration = 1400ms, TR-PCASL = 1420us, 1D spatial encoding perpendicular to the flow, matrix size = 64x1x1 (x,y,z), mean z-gradient = -0.7mT/m, slice distance = 29.5mm, slice thickness = 17.1mm, FOCI pulse for PASL inversion. The phantom had a flow rate of 350ml/min corresponding to about 20.6cm/s of flow velocity and used a perfusate with a T1 of about 1.8s.
Simulation:
An in-house developed Python ASLIF Bloch simulation was used for the model validation. The simulation assumes a 3D laminar flow profile and a straight and static flow between the PCASL and ASLIF excitation planes. Dispersion effects were neglected. The simulation used T2 = 150ms and LEref = 95%. The used T1 was varied between 500-2000ms as well as the maximum laminar flow velocity between 5-70cm/s for different average labeling efficiencies. Figure 3 shows the fit results for the ASLIF simulation together with set reference values of the average labeling efficiency and blood T1. The model fit accurately estimates T1b and the estimated effective labeling efficiency closely matches the average labeling efficiency across the flow distribution with deviations below 5%. Figure 4 shows the model fit together with the measured ASLIF signal for the beginning of a PCASL labeling period on a flow phantom. T1b was set according to the flow phantoms specifications. The fit results show good agreement of the estimates to the set flow velocity and the expected labeling efficiency. The ASLIF model fit accounts for the labeling efficiency, flow velocity and T1b of the PCASL labeled arterial input function. However, the model fit currently does not account for differences in flow dispersion between the PCASL and PASL based ASLIF measurements. Therefore, for the phantom data only the beginning of the ASLIF signal with equal flow dispersion between the PCASL and PASL based ASLIF measurements was used. Due to the short time interval no reliable T1 estimate could be computed. The simulation yields reliable T1b estimates and shows a close correlation of the modeled effective labeling efficiency to the average labeling efficiency and thus its use for respective quantification. A quantification model for the ASLIF signal was created that enables estimates of the labelling efficiency and flow velocity in PCASL measurements. The model was validated with the help of simulations and experiments in the flow phantom. Future plans include in-vivo application and the extension of the fit model to account for different flow dispersion between the ASLIF and the reference measurement for simultaneous T1b estimations.
Luis Andrea HAU (Hamburg, Germany), Thomas LINDNER, Jens FIEHLER, Matthias GÜNTHER
15:40 - 17:10
#47613 - PG433 Impact and correction of irregular heartbeats on inversion-recovery multi-shots cardiac late gadolinium enhancement images.
PG433 Impact and correction of irregular heartbeats on inversion-recovery multi-shots cardiac late gadolinium enhancement images.
Segmented cardiac MRI data acquisitions, such as late gadolinium enhancement (LGE), divide k-space into shots, each shot capturing a portion of the signal at every heartbeat. LGE aims at directly visualizing the infarct lesion through a positive and maximal contrast exploiting the magnetic properties of the tissues, hence relying on magnetization equilibrium between shots to ensure signal consistency across the final k-space. However, irregular cardiac cycles result in variable recovery periods, disrupting this pseudo steady state and undermining image quality and contrast efficiency. Through numerical simulations and acquisitions, this study investigates how irregular heart rates induce artifacts in 2D LGE sequences. Eventually, advanced image reconstruction is proposed to reduce artifacts by substituting corrupted k-space lines as a workaround.
A 2D inversion-recovery segmented FLASH sequence with interleaved reordering and 8 shots (25 segments per shot) was acquired on a 3T MRI scanner (MAGNETOM Cima.X, Siemens Healthineers, Erlangen, Germany) and numerically simulated using the MRzero framework [1] based on Phase Distribution Graph [2]. To best approximate cardiac imaging, the T1MES Phantom was used, with theoretical T1, T2, T2*, B0 and B1+ as provided in [3]. Two conditions were simulated and acquired: a constant and a variable heart rate (HR) (Figure 1). A fixed inversion time (TI) of 250ms was chosen to maximize the contrast between the tubes representing the late post-contrast infarcted (T1=284ms) and the remote myocardium (T1=525ms). The acquisition window was positioned at end-diastole. When one RR interval was reduced, the next trigger occurred within the acquisition window and was missed, prolonging longitudinal T1 recovery for the next shot, thus corrupting corresponding k-space (Figures 1 and 2). Images from regular HR and with one corrupted shot from irregular HR were reconstructed using a simple Fourier transform (FFT). Then the corrupted shot was removed and image was reconstructed using compress sensing (L1-ESPIRiT using BART Toolbox with a L1-wavelet regularization term of 0.03). Images were compared using three metrics: Percent Signal Ghosting [4] (PSG) computed as PSG = | (Top+Bottom) - (Left+Right) | / (2*Signal), the signal enhancement [5] (SE) defined as the relative difference between the late post-contrast infarcted and the normal myocardium, and the normalized mean squared error (NMSE) on the whole image. Irregular ECG events creates artifacts on images that can be significantly reduced by removing the corrupted lines and using compress sensing (Figure 3). The severity depends on the corrupted lines k-space position, with higher ghosting when the center is affected, and more pronounced artifacts as the recovery duration of the corrupted shot grows (Figure 4). The simulations modeled two scenarios: lengthening RR cycles (RR = 1–1.4s), and shortened RR cycles with a missed trigger (RR > 1.5s). This simulation study demonstrates the significant impact of irregular heart rate in segmented MRI sequences with inversion-recovery preparation. Inconsistent timing between shots introduces strong signal fluctuation in the pseudo steady state signal, resulting in signal ghosting and affecting contrast. We show that CS could be a valuable reconstruction candidate to tackle artifacts due to broken equilibrium of the magnetization. Adverse ECG events causing disrupted equilibrium magnetization and k-space inconsistencies can be effectively addressed by identifying corrupted shots and removing the corresponding k-space lines before CS reconstruction. Future work will explore 3D LGE and also the limitations of this approach before applying it to in vitro and in vivo studies and assess its impact on clinical cardiac MRI scans to further optimize image quality and diagnostic reliability.
Cyprien BOUTON (Lyon), Thomas TROALEN, Stanislas RAPACCHI, Pierre CROISILLE, Magalie VIALLON
15:40 - 17:10
#46174 - PG434 Modeling of Direct Saturation from an Off-Resonance Preparation Pulse.
PG434 Modeling of Direct Saturation from an Off-Resonance Preparation Pulse.
In the context of quantitative MRI protocols using sequences prepared with off-resonance radiofrequency (RF) pulses (e.g., Chemical Exchange Saturation Transfer [CEST], Magnetization Transfer [MT]), the usual modeling of the pulse's impact on the on-resonance water component relies on a pure longitudinal magnetization attenuation – also referred to as “direct saturation” – at an exact frequency offset (Δf) [1]. However, this model does not account for the spectral response of the pulse, which is determined by its shape and duration. The residual on-resonance RF response can lead to unpredictable and non-negligible refocusing of the magnetization of the water component, which can become amplified in sequences where RF events repeat within a short delay relative to T2 (short TR, pulse trains), as previously observed in inhomogeneous MT (ihMT) protocols [2] (Figure 1). In this work, we propose and evaluate an advanced model that incorporates this spectral response at the MT-prepared SPGR sequence level in order to predict suitable and unsuitable regimes of direct saturations.
The proposed model is built upon the Extended Phase Graph (EPG) formalism [3] for signals’ simulations, accounting for the various sequence features (RF, spoiling, gradients, relaxation delays) and providing an efficient way to predict any magnetization refocalisation (echoes). It is extended by discretizing the off-resonance preparation pulse into a series of time-constant operators based on the Bloch equations, thus intrinsically taking into account its spectral response across all configuration states. This extension is compared to the usual discrete-frequency saturation model (“Wf”). In order to challenge our model, synthetic signals of a homogeneous phantom (H₂O + NiSO₄ at 3.75 g/L; with T1/T2 = 225/70 ms estimated at 3 T and an apparent diffusion coefficient of 2.2 µm²/ms) are simulated with a preparation pulse (10-ms gaussian pulse, B1peak = 1.95 µT, FWHM = 280 Hz, and Δf = -200 Hz; MTw) and without (MT0) for normalization purpose, while incorporating experimentally estimated B0 and B1+ field inhomogeneities. The signal maps MTw/MT0 are compared to experimental data acquired at 3 T (Siemens MAGNETOM Vida) using the same sequence features (Figure 2) as those in simulation. The common sequence parameters were: TR = 30 ms, TE = 2.0 ms, flip angle (FA) = 5°, ratio of the spoilers’ moment between preparation and readout = 3/1, and no RF spoiling at the readout pulse level. Experimental and synthetic images are shown in Figure 3. The usual (synthetic Wf) model drastically fails to predict the signals’ behavior, and assumes that the on-resonance water component remains mainly unaffected outside regions of high static field inhomogeneities (red arrows; for ΔB0 ≈ -150 Hz, the off-resonance pulse frequency becomes close to the resonance frequency of local spins). Conversely, the stepwise model (synthetic EPG) more faithfully reproduces the experimental image, and succeeds in predicting specific image patterns. The EPG model that discretizes the off-resonance RF pulse shape offers an accurate representation of the NMR signal. This achievement comes with a considerable computational cost due to the recursive nature of the EPG formalism. We proposed an EPG-based model for the characterization of direct saturation in off-resonance pulse-prepared sequences. This framework can be used in quantitative MT and CEST to determine off-resonance frequency ranges for which the Wf model remains valid.
Lucas SOUSTELLE (Marseille), Andreea HERTANU, Maxime GUYE, Guillaume DUHAMEL, Thomas TROALEN, Olivier M. GIRARD
15:40 - 17:10
#47369 - PG435 Open source tool for the measurement and calculation of the Gradient Impulse Response Function.
PG435 Open source tool for the measurement and calculation of the Gradient Impulse Response Function.
Since its inception, the Gradient Impulse Response Function (GIRF) has become a popular tool for characterising gradient system imperfections in MR imaging[1]. Imperfections, such as those caused by eddy currents induced by the gradients themselves, cause the gradient field experienced by the sample to differ from the ideal and requested field. Imperfections lead to artefacts and a degradation of image quality. This is a particular issue in non-cartesian imaging[2], which relies upon fast switching gradients, and the GIRF has been used successfully in reconstruction to reduce these artefacts[3], as well as optimizing pulse design[4]. However, despite wide use cases, there is no end-to-end, open source tool for complete measurement and calculation of the GIRF, with currently available resources either incomplete, out of date, or reliant on field camera hardware[1,5,6]. This work presents a complete, easy to use tool for the measurement and calculation of the self and B0-cross terms of the GIRF which does not rely on additional hardware. Code for sequence generation, data processing, and GIRF creation is included in a single open-source repository.
The implemented GIRF measurement is based on the optimized method introduced by Robison[7] using positive and negative slice offsets and triangular gradient blips. Additionally, by default, 5x5 2D phase encoding[8] is used with the thin slice method to reduce T2* decay. This allows for longer sampling of any long-time-constant eddy currents (out to about 400 ms), improving spectral resolution of the GIRF. Other parameters including the slice offset, slice thickness, fov, and the number of triangular gradients are user configurable, but by default, these parameters follow the protocol introduced by Wu et al[5]. This method implements interleaved reference scans, with the scheme also following the protocol of Wu et al[5]. The sequence is implemented using Pypulseq[9] to be agnostic to the scanner vendor or version.
The calculation of the GIRF is performed in Python and beyond minor improvements and modifications to account for the phase encoding steps, it is a translation of the Matlab code provided by Wu et al[5] in the optimized calculation. Both the self term (e.g. the effect of the x gradient on x) and the B0 cross terms (e.g. the effect of the x gradient on B0) of the GIRF are calculated. The tool lets the user choose how much of the ADC readout to use, allowing them to decide the balance of spectral resolution and SNR.
All of the code for the sequence, data processing and GIRF calculation are available at https://github.com/jbbacon/GIRF_PE_Python. The code is written in Python and has complete terminal integration. The package manager, Pixi, may be installed by the user allowing them to set up an environment from which the tool can be used. The code for sequence generation and data processing is made fully open-source.
As an example of its use, the GIRF on a 3T Siemens Prisma scanner and a Siemens Magnetom 7T Plus were measured. The default sequence parameters were used with a spherical oil phantom to measure and calculate the GIRF. The magnitude of the self and cross terms of the GIRF with slice offsets in the ‘z’ gradient direction are displayed in Figures 1 and 2 using an ADC readout time of 100 ms during calculation. The ‘x’ and ‘y’ gradient direction terms were also measured and calculated.
The processed data from these examples can be found at https://zenodo.org/records/15352984. The tool was successfully used across multiple scanners to measure and calculate the self and B0 cross terms of the GIRF. The tool used optimized protocols and 2D phase encoding to extend the ADC readout time allowing for improved spectral resolution.
The code has only been tested on data from Siemens VE-line scanners, and as such, the processing of raw data acquired from the scanner is only possible for Siemens data files. We hope to broaden the examples in the future.
We also plan to implement calculation of linear cross-terms (e.g. the effect of the x-gradient on y). This study introduces an end-to-end, open source tool to measure and calculate the GIRF. The tool is easy to use with full terminal integration and makes use of optimized protocols and 2D phase encoding.
James B BACON (Oxford, United Kingdom), Peter JEZZARD, William T CLARKE
15:40 - 17:10
#46396 - PG436 Off-Center Eddy Current Correction for Improved Fat Suppression in Shoulder FSE.
PG436 Off-Center Eddy Current Correction for Improved Fat Suppression in Shoulder FSE.
Eddy currents are a well know source of gradient imperfections that can lead to a variety of imaging artifacts. Therefore, eddy current correction (ECC) by gradient pre-emphasis [1] is implemented on all modern MR systems. It is less well-known that eddy current related magnetic fields often manifest in spatially higher orders [2,3]. This becomes relevant when performing imaging at off-center regions, such as in shoulder MRI. Here, commonly applied spectral fat-suppression can fail due to the off-resonance created by higher-order eddy currents.
To address this issue, we developed an off-center optimized 0th and 1st order ECC and applied it to fat-suppressed FSE shoulder imaging.
Field Camera Measurements
To measure the spatio-temporal eddy current responses, the field evolution after playing out a long trapezoid gradient in x,y and z direction were recorded using a dynamic field camera (DFC) by repeating the measurement multiple times with shifted delays and by placing the DFC at multiple positions along the x-Axis to cover all off-center imaging regions.
Phantom Measurements
In addition to the DFC measurements, a time-resolved (multi-echo) FFE readout following a long trapezoid gradient was performed on phantoms of x:45 cm × y:15 cm × z:15 cm. The magnetic field information was extracted from the phase information of the phantom for each echo.
Processing
From the measured fields, eddy current correction parameters that parameterize the multi-exponential pre-emphasis ECC kernel were calculated by minimizing the difference of the measured fields and the simulation of the ECC fields, which were calculated by applying the ECC kernel to the nominal gradient input.
In-vivo Imaging
Subsequently fat-suppressed shoulder FSE was performed at 20 cm off-center in the x-direction with regular iso-center optimized ECC (optimized for imaging at iso-center) and the proposed off-center optimized ECC. To distinguish ECC effects from potential static B0 effects, imaging was performed twice, once with phase encoding direction in AP and once with phase encoding in PA direction. Eddy currents and off-center optimized ECC
Figure 1 shows an example of an eddy current response following the trapezoid input gradient lobe in x, y, and z direction respectively. When using regular (iso-center optimized) ECC (Fig. 1 left), off-resonance did not show visible gradients in the iso-center, indicating that iso-center optimized ECC is working well. However, in regions being off-center in x direction, substantial off-resonance gradients are visible. When applying simulated off-center optimized ECC for a target x-off-center of 20 cm, where shoulder imaging is typically performed, the eddy current gradients in this region can be significantly reduced (Fig 1, right).
When quantifying the eddy current related field off-set and gradients over time (Fig. 2), applying regular (iso-center optimized) ECC (Fig. 2a) results in off-resonance of over 100 Hz and several hundred Hz/m off-resonance gradients in off-center regions. These can be strongly diminished when using the proposed off-center optimized ECC (Fig 2b). Phantom-based measurements: Initial phantom-based experiments showed a strong discrepancy against the DFC based field measurements. By comparing with the DFC measurements, it was possible to identify that the phantom-based field measurements were sensitive to static B0 off-resonance. After adjusting the sequence and processing to diminish these effects, the phantom-based and DFC-based ECC simulations showed similar behavior (Fig. 2b,c). Some remaining differences (Fig. 2b,c) are observed during the period directly after the gradient lobe where the simulated ECC based on the phantom data was exceeding 15 Hz and 100 Hz/m respectively, possibly due to field echoes transitioning into steady-state. Errors of such magnitude however may not strongly effect spectral fat suppression and thus may be acceptable.
In-vivo imaging (Fig. 3): For the images with regular (iso-center optimized) ECC, incomplete fat suppression and local signal reduction are visible. These effects changed when switching the phase encoding direction from AP to PA, indicating that the reason for the artifacts is incomplete ECC. These effects were strongly diminished, and the images with non-switched and switched PE-direction were more similar to each other, when applying off-center optimized ECC. We developed a method to achieve off-center optimized 0th and 1st order ECC, which strongly diminished eddy currents for imaging in a targeted off-center region, and demonstrated improved fat suppression in shoulder FSE. In addition, we showed that it was possible to develop and validate a phantom-based ECC using field camera measurements as a reference.
Bertram WILM (Zurich, Switzerland), Ryohei TAKAYANAGI, Yuki SAKATA, Masaaki UMEDA
15:40 - 17:10
#46304 - PG437 Robust variable flip angle T1 mapping for imperfect field conditions.
PG437 Robust variable flip angle T1 mapping for imperfect field conditions.
Quantitative MRI (qMRI) focuses on the direct measurement of tissue parameters and composition [1-4]. Absolute measurements of tissue composition facilitates reliable comparisons across subjects and imaging systems [3,5,6]. Among qMRI parameters, the longitudinal relaxation time T1 is widely used in assessing various pathologies, including brain diseases, iron overload, cancer, and cardiac disorders [7]. However, T1 values vary substantially across methods and systems [8].
Variable flip angle (VFA) techniques allow fast 3D T1 mapping but are sensitive to radio frequency (RF) field (e.g. B1) inhomogeneities, especially at high fields. Incorporating B1 maps into post-processing improves accuracy, but increases scan time and complexity [9]. As well, it adds the possibility of B1-map introduced errors. Robust excitation pulses that reduce B1 and B0 dependence could improve VFA-T1 mapping accuracy. We present RF pulses by optimal control (OC) meeting these requirements and demonstrate improved accuracy in simulations and phantom experiments on a preclinical 7T system.
The core of this work is a pair of excitation pulses designed by optimal control with robustness to imperfections in the B0 and B1 field [10,11]. Optimization objectives were:
• Non-selective excitation with flip angles 6° and 20°.
• B1 robustness for a range of 75% to 120% scale of the nominal amplitude (50 μT).
• B0 robustness for -4 to 4 ppm, assuming a field strength of 7T.
• Short pulse duration.
To compare VFA based on OC with commonly used RF pulses, we designed two block pulses with the same flip angles and amplitudes (50 μT) as OC. VFA T1 mapping is then simulated using the full Bloch equations, solved with symmetric operator splitting [12], over a broad range of field imperfections, and assuming relaxation times T1 = 2000 ms and T2 = 500 ms.
We implemented the optimized RF pulses and the block RF pulses into a VFA T1 mapping protocol on a Bruker BioSpec 70/30 (Bruker, Ettlingen, Germany). Two 3D non-selective spoiled gradient echo acquisitions were acquired (flip angles 6° and 20°, TE = 6.23 ms, TR = 15 ms, acq. matrix 80 x 128 x 128). To obtain ground-truth T1, a single voxel inversion recovery experiment with 25 inversion delays was acquired. Phantom scans were performed using a syringe filled with 5ml of tap water. Figure 1 illustrates the numerical prediction of T1, with a nominal T1 value of 2000 ms for a wide range of B1 scales and resonance offsets. OC pulses (Fig. 1A) outperformed block pulses (Fig. 1B) in terms of T1 accuracy within the optimization target area, particularly for B1 scales deviating from the nominal value of 100%. Figure 1C further highlights the improved uniformity of OC pulses with respect to B1 scales for no off-resonance. In contrast, block pulses exhibit quadratic dependence on the B1 scale (fit not shown).
Inversion recovery measurement of the phantom resulted in a nominal T1 of 1915 ms (not shown). The T1 predicition with VFA in phantom (Figure 2) demonstrates that block is more homogeneous within the entire phantom, but severely underestimates T1. In contrast, with OC we have a more accurate depiction of T1 compared to the nominal value.
Table 1 summarizes the analysis of predicted T1 values based on simulation with the full Bloch equations within the optimization target area (red box in Figure 1). The minimum and maximum T1 values obtained with OC pulses are substantially closer to the nominal value than block pulses. The 90% range with OC pulses shows less than 15% deviation of the nominal value, while with block pulses this deviation exceeds 40%. Optimal control excitation pulses with robustness to field imperfections were applied to variable flip angle T1 mapping. OC pulses enabled T1-prediction accuracy surpassing block RF pulses, in particular in the presence of B1 imperfections. With block pulses, the dependence on the B1 scale is quadratic. Prior methods introduced a quadratic correction term [9] that requires acquisition of an additional B1 map. However, the accuracy of the T1 map is then tied to the accuracy of the B1 map. In addition, this approach prolongs acquisition time.
The proposed OC pulses do not require B1 maps within post-processing, reducing scan time and potential inaccuracies. The logical next step is applying optimal control excitation pulses for VFA T1 mapping in human in vivo MRI at 3T and 7T. The advancement in VFA T1 mapping using optimal control excitation pulses reduces scan time since no additional B1 map is required, and it minimizes potential errors due to inaccuracies in the B1 maps and their correction.
Christina GRAF (Vancouver, Canada), Alexander JAFFRAY, Clemens DIWOKY, Armin RUND, Stefan STEINERBERGER, Alexander RAUSCHER
15:40 - 17:10
#46452 - PG438 Assessment of motion impact on non-parametrized dynamic pTx pulse performance at 7T.
PG438 Assessment of motion impact on non-parametrized dynamic pTx pulse performance at 7T.
Dynamic parallel transmission (pTx) has shown promise in mitigating flip angle (FA) nonuniformity, a major challenge at ultra-high field strengths due to severe B1+ inhomogeneity [1]. Recent developments now enable fast online customization (FOCUS) of dynamic pTx pulses by using low resolution B0 and multi-channel B1+ maps to optimize a subject-specific dynamic pulse [2]. This method yields more uniform FA distribution compared to static pTx [3] or universal pulses [4], but it adds additional acquisition time (approximately 1 min) prior to the scan. Because of this significant preparation time, the maps are typically acquired only once at the beginning of the session and then reused for the pTx pulse optimization of all sequences in the session. However, any subject motion occurring after the initial mapping can change the B0 and B1+ distribution, and, in turn, degrade pulse accuracy [5].
Building on our earlier evaluation of motion-induced effects on FA distributions using kT-point based pulses [6,7], here we extend the methodology to non-parametric dynamic pTx pulses recently introduced to enable FOCUS pTx in SPACE applications requiring variable high FAs [8]. By enforcing symmetric RF and anti-symmetric gradient shapes, this pulse can be scaled to a wide range of FA by varying the transmission voltage [9]. In this study, we aim to assess their performance when subject motion occurs after the acquisition of B₀ and B1⁺ maps. Furthermore, as rigid-body registration effectively approximates small motion-induced B0 changes [10], we explore the use of spatially registered maps from the original head position to the new position to re-optimize the pulse, thereby eliminating the need to re-acquire field maps.
Eleven healthy volunteers (5 females, age range = [24-41] y/o) were scanned at 7T (MAGNETOM Terra.X, Siemens Healthineers, Forchheim, Germany) using an 8Tx/32Rx RF head coil (Nova Medical, Wilmington, USA). A multi-echo GRE (res=4.0×4.0×6.0mm3, TEs=[2.39,4.59,7.09]ms, TR=10ms, TA=12s) and a pre-saturated TurboFLASH (res=4.0×4.0×5.0mm3, TR=3780ms, TA=45s) sequence were acquired at 15 different head positions to compute B0 and B1+ maps, respectively [11,12].
As described in Figure 1, three different pTx pulses were optimized for a target FA of 90° for each head position (i):
1. Motion: the first pulse was optimized on the maps acquired at the first position, representing the case where there has been a motion between two acquisitions but the field maps have not been re-acquired;
2. No motion: the second pulse was optimized on the maps acquired at position i, thus representing the case where there is no motion between the acquisition and the optimization;
3. Motion + registration: the last pulse was optimized with B0 and B1+ maps from the first position registered to position i.
Resulting FA maps were then computed using Bloch simulations for the three pulses at each position [2]. FA map homogeneity was evaluated with the mean relative error (MRE) with respect to the target (flat 90° map) and pulses were compared pairwise using the Wilcoxon signed-rank test. Between the first position and all the others, the framewise displacement (FD) was computed to measure motion amplitude [13]. Example of resulting FA maps at one head position are shown in Figure 2. The “no motion” pulse results in a flatter FA map than the other two pulses. FA maps from the “motion” and “motion + registration” pulses exhibit similar FA distribution.
Figure 3 shows the distribution of MRE for all subjects and positions. A significant difference was found between the MRE of the “no motion” pulses and both the “motion” and “motion + registration” pulses (p<1×10-24) with a median MRE increase of 0.72% and 0.85%, respectively.
Figure 4 presents the MRE of the “motion” pulses against the FD. A significant positive correlation (ρ=0.53, p=1.5e-12) was observed between the MRE and the FD. However, most points are clustered by subject and the correlation is not consistent in individual subjects. When tested separately, only 2 subjects out of 11 showed significant correlation (after Bonferroni correction for multiple comparisons) between MRE and FD. This work demonstrates that non-parametrized pTx pulses optimized using the FOCUS framework exhibit robustness to motion in the range we measured (FD < 27.6mm). The resulting median increase in MRE when not accounting for motion was only 0.72% over the whole brain, much smaller than the MRE of the pulses themselves compared to the target 90°. Results suggest that the motion-induced increase in FA map inhomogeneity is not correlated with the amplitude of the movement but appears to be subject-dependent. Finally, registering the maps to the new head position yielded no improvement over the “no motion” pulses. These results indicate that FOCUS-optimized pTx pulses can maintain acceptable FA homogeneity in presence of motion, supporting their practical application in real-time or motion-prone settings.
Jocelyn PHILIPPE (Lausanne, Switzerland), Natalia PATO MONTEMAYOR, Antoine KLAUSER, Emilie SLEIGHT, Patrick LIEBIG, Jürgen HERRLER, Robin HEIDEMANN, Jean-Philippe THIRAN, Tom HILBERT, Gian Franco PIREDDA, Thomas YU
15:40 - 17:10
#47820 - PG439 A study of the effect of chemical shift displacement artifact on Quantitative Susceptibility Mapping in the breast.
PG439 A study of the effect of chemical shift displacement artifact on Quantitative Susceptibility Mapping in the breast.
Breast cancer is a leading cause of death in women worldwide[1]. A common marker of malignancy is breast calcifications[2] detected by X-ray mammography, a procedure that involves ionizing radiation and has low sensitivity in dense breasts[3]. Breast MRI allows microcalcifications to be identified in gradient echo (GRE) phase data and Quantitative Susceptibility Mapping (QSM). It has also shown high sensitivity to the diamagnetic properties of microcalcifications[4,5], flagging a potential role in their identification.
QSM estimates tissue magnetic susceptibility from MR image phase[6]. While it offers excellent contrast in brain imaging, breast QSM faces additional challenges due to fat. The different resonance frequencies of water and fat cause chemical shift artifacts (CSA), including fat displacement at low bandwidths (type-1 CSA) and phase cancellation in GRE acquisitions (type-2 CSA)[7]. Strategies to address these effects include effective in-phase acquisitions (EIP)[8] and Simultaneous Multiple Resonance Frequency (SMURF) imaging[9]. EIP acquisitions use corrected TEs based on the 6-peak fat model to reduce type-2 CSA. SMURF uses multiband pulses to excite water and fat separately but simultaneously, enabling correction of both CSA types before recombining signals. These corrections are especially valuable at low bandwidths, where SNR is higher but type-1 CSA is more severe.
In this work we evaluated the effects of type-1 CSA on QSM estimations in breast imaging. To do so, we used as ground truth QSM maps of the breast obtained from SMURF reconstructions (i.e, no displacement CSA), and compared them with QSM estimations obtained with effective echo time imaging over a wide range of bandwidths.
We acquired two coronal 3D monopolar mGRE sets from a healthy female volunteer on a 3T scanner (Siemens PRISMA) using the 28-channel BraCoil array[10]. The first dataset was acquired using effective in-phase time-interleaved echoes, using the bone marrow model TEs[8]. Acquisition parameters were: matrix size=432x208x160, resolution=0.9x0.9x1.2mm, TE={2.38,4.59,6.81,9.17}ms, TR=9.6ms, rBW/px=500Hz/px, flip angle=10°. The second dataset was acquired using SMURF imaging, with RL phase-encoding direction. Acquisition parameters were: matrix size=360x208x160, resolution=0.9x0.9x1.2mm, TE={4.96,12}ms, TR=24ms, rBW/px=400Hz/px, flip angle =10°.
To quantify the effects of type-1 CSA, we corrected the 400 Hz bandwidth fat displacement on SMURF data and re-shifted the fat images simulating 200, 400, 800, 1200, and 1600Hz/px acquisitions. Then, type-2 CSA and T1 corrections were applied to the different SMURF images. Both EIP and SMURF datasets were pre-processed using ROMEO[11] unwrapping and PDF[12] background field removal. QSM maps were estimated using FANSI[13]. Reconstruction scores were computed using normalized RMSE and XSIM[14], using the fully-corrected SMURF acquisition as ground truth. Figure 2 shows the images resulting from EIP and SMURF acquisitions. In both cases, fat displacement artifacts of 0.86 px and 1.08 px are present due to the respective 500 and 400 Hz/px receiver bandwidth of EIP and SMURF.
Figure 3 shows the simulated fat displacements after applying Type-2 CSA, T1–bias corrections and coil combination. At high bandwidths (e.g., 1200–1600 Hz/px), Type-1 CSA effects are minimized and images resemble the reference. Low bandwidths show 1-2 px fat shifts, seen as signal voids in ROIs.
Figure 4 shows QSM reconstructions and their differences from the fully corrected SMURF images. As fat displacements decrease, streaking artifacts are reduced and XSIM improves, with slower gains beyond 1000 Hz/px. At 200 Hz/px, strong streaking appears near mammary glands due to signal voids. Fat displacement artifacts depend strongly on receiver bandwidth. While EIP cannot correct them without increasing bandwidth, SMURF enables mitigation through Type-1/2 CSA and T1–bias correction. As bandwidth increases, fat shifts decrease, resulting in more accurate images and improved similarity to the reference. In contrast, low bandwidths lead to 1-2px signal voids in fat-rich regions, degrading image quality. QSM difference maps confirm that reducing displacement improves XSIM metrics, although XSIM improvements become less pronounced above 1000 Hz/px. At very low bandwidths, unrecoverable signal loss causes severe streaking artifacts, particularly around the mammary glands. We simulated and quantified the effect of the fat displacement artifact on breast QSM at different receiver bandwidths. Our results show that the signal voids at bandwidths under 800 Hz/px can severely impact susceptibility estimations if not corrected, especially in high tissue variability regions. This consideration is particularly relevant for approaches like in-phase acquisitions, which are not able to correct the type-1 CSA.
Javier SILVA (Chile, Chile), Beata BACHRATA, Carlos MILOVIC, Simon Daniel ROBINSON, Cristian TEJOS
15:40 - 17:10
#47946 - PG440 Explaining motion artefacts in 3D neuroimaging MRI using motion-sampling plots.
PG440 Explaining motion artefacts in 3D neuroimaging MRI using motion-sampling plots.
Motion during magnetic resonance imaging (MRI) is a well-known source of image artefacts, particularly in 3D, where data acquisition times are long1. Different portions of k-space are acquired at different time points, and if the subject moves the sampled data can represent different poses of the anatomy. This leads to inconsistencies across k-space that ultimately manifest as artefacts in the reconstructed image.
However, many experimental studies investigating motion-induced artefacts define motion purely as a function of time2-4, with limited attention to how motion interacts with the k-space sampling trajectory. This can obscure understanding of why certain artefacts appear as they do. To address this, we propose the use of motion-sampling plots5—a method of visualizing the joint relationship between motion states and k-space locations during data acquisition.
Motion-sampling plots offer a more intuitive understanding of how specific artefacts arise from specific combinations of motion and sampling. In this study, we demonstrate how motion-sampling plots can enhance the interpretation and communication of motion artefacts in both simulations and in-vivo experiments.
We conducted a series of simulations and in-vivo experiments to investigate the effects of head motion during 3D MRI acquisitions. Here we focus on nodding motion (i.e., rotation around the left–right axis) as a representative and commonly encountered type of rigid-body movement in neuroimaging.
We analyzed how this motion interacts with different sampling strategies: (1) Cartesian sampling, (2) stack-of-stars sampling, and (3) kooshball sampling. For each sampling method, we further considered both smooth and non-smooth view ordering to examine its effect on motion artefacts.
Each motion trajectory was plotted in a motion-sampling space, which maps motion state against corresponding k-space location. The resulting artefacts in reconstructed images were categorized and linked to features visible in these plots.
In addition, we analyzed five real motion trajectories recorded from patients during scanning and studied their impact across the various sampling strategies on healthy volunteers. Our results demonstrate that the same set of motion states, when applied to different parts of k-space, can produce artefacts that differ markedly. This underscores the need to consider not just how much a subject moves, but during which part of the sampling the motion occurs (Figure 1).
We identified and categorized distinct artefact types, each associated with specific motion-sampling features (Figure 2):
1. Superposition occurs when central k-space is sampled in multiple motion states.
2. Ghosting results from stripy distributions in Cartesian sampling.
3. Noiselike artefacts appear when motion states are randomly scattered.
4a–b Ringing arises from discontinuities parallel to the imaging plane (Cartesian) or conical wedges in k-space parallel to the imaging plane (Kooshball).
5a–b Lines emerge from a central discontinuity in the imaging plane in both Cartesian and stack-of-stars sampling.
6. Streaking occurs with distinct wedges in the imaging plane (radial schemes).
7. Edge wobbles reflect smooth pose drift across k-space.
We also observed increased artefact severity with greater motion amplitudes, including features suggestive of potential secondary effects such as motion-induced B0 shifts (Figure 3).
In real patient motion trajectories, we observed that the same movement patterns yielded dramatically different image artefacts depending on the sampling scheme, and these differences were interpretable through motion-sampling plots (Figure 4). This study is focused on rigid-body nodding motion, which represents a simplified yet relevant subset of head motion. While this limits generalizability to more complex motion types, we see similarities in artefacts and motion-sampling interactions in the real world motion cases.
Our findings suggest that many commonly observed motion artefacts can be traced back to identifiable motion-sampling interactions. This framework offers a more k-space-centric perspective that can better explain image degradation in moving subjects. We have shown that the relationship between motion and sampling trajectory—captured via motion-sampling plots—is critical when describing and interpreting motion artefacts in 3D MRI.
By accounting for where in k-space motion occurs, motion-sampling plots offer insight into the mechanisms of artefact formation, reveal why certain motions cause more severe degradation, and explain variability across sampling strategies. This approach enhances reproducibility and clarity in motion studies and has the potential to guide both future research and practical acquisition design.
Ultimately, to improve motion robustness in MRI, it is essential not only to quantify motion but to understand how motion and sampling jointly shape the image.
Sophie SCHAUMAN (Stockholm, Sweden), Adam VAN NIEKERK, Henric RYDÉN, Ola NORBECK, Stefan SKARE
15:40 - 17:10
#46313 - PG441 Rapid and low-cost evaluation of shielded room effectiveness in clinical MRI with a software defined radio.
PG441 Rapid and low-cost evaluation of shielded room effectiveness in clinical MRI with a software defined radio.
In magnetic resonance imaging (MRI), images are produced from a tiny radiofrequency (RF) signal. To prevent this signal from being altered by RF interference, an RF shielding enclosure is required[1]. Typically, the RF enclosure is built before the MRI system is installed, and often many MRI system upgrades or replacements are carried out in the same shielding room. The ageing of shielding room components must therefore be taken into account. In particular, the sealing system of the doors, which are opened and closed several times an hour, is susceptible to damage. Long-term effectiveness must therefore be assured and measured periodically. A radio frequency signal RF1 generated at the frequency of interest is measured through the shielding at a value RF2, the shielding efficiency is expressed in dB as 20 log(RF1/RF2) representing the attenuation obtained by the shielding. These periodic measurements are costly because they are carried out by experts equipped with expensive control systems (RF generator and network analyser)[2]. The aim of the present study was to propose an experimental set-up consisting of an SDR device and software to measure the same RF MRI pulse sequence generated by the MRI both inside and outside the shielding room, enabling simple evaluation of shielding effectiveness.
To illustrate the method, a schematic representation is given in Fig. 1. First, a 40 mm diameter RF coil matched to 50 ohms with a very low quality factor to enable broadband reception is manufactured. This receiver coil is placed at various positions along the walls inside the shielding room, then the same measurements are made at the corresponding positions outside the room (Fig. 2). This coil is connected to the input of an SDR dongle (Nooelec, USA) via fixed-value RF attenuators (Mini-Circuits, USA) to take account of the huge variations expected in signal amplitude between outdoor and indoor measurements. This dongle is plugged into a computer's USB port, and the open source CUBICSDR software (https://cubicsdr.com/) is played on the computer. The SDR frequency is set to MR frequency and In-Phase and Quadrature (I/Q) mode is selected with a receive bandwidth of 192 kHz. Frequency and time windows allow to view signals and adjust both the SDR tuner gain and/or the fixed attenuators. The demodulated signal is recorded in a wav file at the frequency of the receiving bandwidth. The wav file is then decoded using an in-house developed Mathematica (Wolfram Research, Inc., USA) script. This script provides temporal and frequency displays of selected parts of the sequence of interest (Fig. 3). This script enables precise measurement of the durations, relative amplitudes, and frequencies of recorded pulses[3]. To illustrate the capabilities of the set-up, three shielding room were evaluated, two enclosing a 1.5 T clinical MRI at 63.64 MHz (Magnetom Sola, Siemens Healthineers, Germany) and the third a 3T systems (Magnetom Vida, Siemens Healthineers, Germany). 2D gradient echo sequences were played repeatedly on the systems and measured at different positions (Fig. 2). An additional measurement was made for both two systems with the door slightly open, to check the validity of the method. The Table 1 shows the measured values of the signal attenuation (in dB) by the shielding rooms. Shielded Room 3 has recently been renovated with a brand-new door, unlike the other two which have been in place for several years. We observe that the attenuation of this room is higher (85 dB) than the two older shields, which is consistent, as shields degrade over time, mainly caused by door aging. Furthermore, when the door is slightly open, the attenuation is lower than when it is closed, indicating, as expected, a greater RF leakage. All this Results therefore confirms the reliability of the method. The low-cost method we propose works at both low and high fields, and could therefore be used for inexpensive control of isolation in many MRI systems. However, this method cannot be used to certify an RF shield, as the IEEE 299-2006 « Standard Method for Measuring the Effectiveness of Electromagnetic Shielding Enclosures » is not precisely followed. We designed an experimental setup consisting of a home-made broadband RF coil connected to a software-defined radio (SDR) to measure the same pulse sequence produced by the MRI, both inside and outside the shielding room, allowing direct assessment of shielding effectiveness. This simple, low-cost method can be used for periodic checking of the room efficiency without the need for an expert and his complex equipment (RF generator and network analyser).
Kouame Ferdinand KOUAKOU (Angers), Anita PAISANT, Vanessa BRUN, Christophe AUBE, Hervé SAINT-JALMES
15:40 - 17:10
#47876 - PG442 Linewidth Analysis of MR Spectrum in AI-Based Dynamic Shimming in the Presence of Motion.
PG442 Linewidth Analysis of MR Spectrum in AI-Based Dynamic Shimming in the Presence of Motion.
For high-quality MR imaging, it is essential that the main magnetic field (B₀) remains homogeneous throughout the acquisition[1]. Inhomogeneities in the B₀ field impact magnetic resonance spectroscopy (MRS) by causing spectral line broadening[2]. Shimming with spherical harmonics (SPH) coils is the primary approach to cancel these inhomogeneities[3]. An array of circular AC/DC coils provides superior shimming performance compared to the conventional spherical harmonics (SPH) coils integrated into the MR scanner[4].
Conventional static shimming applies B₀ correction only before the scan, whereas dynamic shimming continuously updates B₀ throughout the scan[5]. shimming updates are often based on the initial GRE B₀ pre-scan, or they measure B0 changes continuously[6]. Nevertheless, reliably measuring these B₀ changes remains a challenging task.
Volumetric navigators, interleaved with the primary acquisition, provide a means to dynamically track B₀ changes throughout the scan[7], [8]. They increase the overall scan time if the primary sequence lacks adequate dead time[9]. Motyka et al. proposed a deep learning (DL) based method for predicting motion-induced B₀ inhomogeneities, which has shown comparable results to that of EPI-based dynamic shimming[10].
Linewidth measuring through full width at half maximum (FWHM), are commonly used to assess MRS quality[11]. In this study, we assess the effects of motion-related B₀ inhomogeneities on a simulated water FID signal. We then evaluated the FWHM of its Fourier-transformed frequency spectrum under various shimming conditions (static vs. dynamic) and across two coil configurations: (i) SPH and (ii) SPH+AC/DC.
This study uses a dataset from Motyka et al., involving 15 healthy volunteers scanned using a 7T Magnetom+ MR scanner (Siemens Healthineers, Erlangen, Germany)[10]. For each subject, multi-echo B₀ maps were acquired using 2D GRE and 3D EPI sequences at 30 randomly selected head positions (1 initial and 29 after-motion). Additionally, Anatomical MP2RAGE images were acquired at the initial position and then transformed to the other positions using transformation matrices generated from the co-registration of the 2D GRE magnitude images[10].
To generate a semi-synthetic dataset shimmed with SPH and AC/DC coils in static mode, the original dataset was shimmed with 31-channel AC/DC coil fields in two regimes: (i): Whole-brain (ii): Slice-specific (Figure 1).
A DL neural network based on U-Net with subject-specific fine-tuning was trained using the Static-SPH+AC/DC dataset in two regimes: (i): Whole-brain (ii): Slice-wise. The network inputs included anatomical images at both the initial and after-motion positions, as well as the B₀ field maps at the initial position, with the goal of predicting B₀ maps at the after-motion positions. Training was carried out on data from 11 participants across 30 head positions. For evaluation, data from the rest 4 participants were used, where the first 6 after-motion positions were reserved for subject-specific fine-tuning and the remaining 23 positions for testing.
Dynamic shimming with SPH+AC/DC coils was simulated by calculating shim field maps for each after-motion position, based on DL-predicted and measured GRE and EPI based B₀ field maps, and subtracting them from the B₀ maps of corresponded positions (Figure 1).
To evaluate the efficiency of DL-based dynamic shimming on MRS, we simulated the FID signal of water and applied B₀ inhomogeneities. The FWHM of the Fourier-transformed spectrum at each voxel was calculated. Finally, the mean and standard deviation (STD) of the FWHM across all voxels for each shimming method were computed, along with the percentage of inappropriate voxels (FWHM > 0.1 ppm)[11]. A post-hoc test was then conducted on all results. AC/DC coils helped to make the magnetic field more homogeneous when the subject was still, but movement reduced this. However, dynamic shimming preserves the homogeneity (Figure 2).
For the DL-based dynamic shimming method, STD of FWHM measurements is significantly lower than that of the EPI-based method, while the mean shows similar results (Figure 3).
The percentage of inappropriate voxels didn’t show any significant difference between DL-based dynamic shimming and those with GRE and EPI based methods (Figure 4). Although AC/DC coils improve initial B0 homogeneity, motion degrades it, highlighting the need for dynamic shimming. The proposed DL-based method matches GRE and EPI dynamic shimming and may replace volumetric navigators by enabling rapid, high-resolution shim updates. This simulation study demonstrates the potential of DL networks for real-time shimming in the future. This study assessed the FWHM of simulated FID signal of water with applied motion-related inhomogeneities under different static and dynamic shimming using additional AC/DC shim coils. DL-based dynamic shimming performed similarly to GRE and EPI based approaches, but with no added scan time.
Mohammad KHOSRAVI (Klagenfurt, Austria), Wolfgang BOGNER, Bernhard STRASSER, Jason STOCKMANN, Günther GRABNER, Beata BACHRATA, Stanislav MOTYKA
15:40 - 17:10
#47900 - PG443 3d radial 4d flow mri in a cardiorespiratory motion flow phantom with eddy current artifact correction.
PG443 3d radial 4d flow mri in a cardiorespiratory motion flow phantom with eddy current artifact correction.
4D flow MRI is a technique that provides measurement of blood flow in three spatial dimensions over time, allowing for detection of abnormalities in the cardiovascular system [1]. However, due to the four-fold increase of required readouts compared to anatomical scan sequences, a compromise must be made between scanning times and imaging artifacts caused by undersampling.
3D radial k-space trajectories sample the center of k-space at every repetition, providing less sensitivity to motion and undersampling artifacts compared to Cartesian strategies [2]. However, due to rapid switching of gradients, eddy current artifacts such as gradient delays and B0 phase errors must be addressed [3]. This work presents the investigation of two 3D radial trajectories for 4D flow MRI with eddy current artifact correction in a pulsatile flow phantom with cardiorespiratory motion.
Two 3D radial trajectories (shown in figure 1) were adapted from literature and further developed for application on a 3T Philips MR7700 scanner:
Golden means (GM), where the two-dimensional golden ratios ϕ_1=0.4656 and ϕ_2=0.6823 are used to calculate the azimuthal and polar angles of each 3D radial spoke [4].
Spiral phyllotaxis (SP), with successive readout spokes spiraling down in an interleaved fashion. Each interleaf is rotated by the golden angle of 137.5° from the preceding one [5].
Data acquisition was performed on a pulsatile flow phantom mimicking both aortic and respiratory motion (LifeTec Group, Eindhoven, The Netherlands). The moving phantom was programmed to pump water through a flexible tube with downstream resistance at a fixed rate of 60 bpm, while mimicking respiratory motion in the head-foot direction at a fixed rate of 10 bpm (shown in figure 2).
4D flow measurements were conducted for both radial trajectories with the following scan parameters: TR/TE = 4.08/1.66 ms, α = 4.5°, venc = 100 cm/s, FOV = 256mm, isotropic resolution = (1.33 mm)3, acquisition time ≈ 10 min, number of readout spokes ≈ 36,600. One scan was acquired per trajectory. Image reconstruction was performed in Matlab (R2022b; MathWorks) using a parallel-imaging and compressed-sensing algorithm, which provided NUFFT gridding and density compensation [6]. The reconstruction algorithm was combined with retrospective binning to reconstruct end-expiration 4D flow images with 10 cardiac phases.
To measure and correct eddy current-induced artifacts, separate 15-second calibration GRE scans with no motion were performed with the same scan parameters and parts of the trajectory as the 4D flow measurements. These datasets were used to calculate gradient delays using a RING method from BART [5] and to correct B0 phase errors with an adapted 3D version of a previous method [3], using to the following equation:
ϕ_ec (ϕ,θ)=ψ_x G_x cos(ϕ) sin(θ)+ψ_y G_y sin(ϕ) sin(θ)+ψ_z G_z cos(θ)+ϕ_0
Here, ϕ_ec is the accumulated B0 phase error as a result of the azimuthal (ϕ) and polar (θ) angle of the readout spokes and gradient strengths G. The phase errors per gradient strength ψ and the constant phase offset ϕ_0 were fitted for each coil using a non-linear least squares fit. The fits were applied to the 4D flow datasets as a phase shift to the raw k-space data. Only coils with a good fit (R^2≥0.8) were used for reconstruction, after further compressing the coils down to 8 virtual channels to reduce the computational burden. Gradient delay trajectory compensation was found to be minimal in all directions for both trajectories, with the largest compensation being 0.45 voxels in the AP-direction at an isotropic k-space FOV of 130 voxels. After removing the constant phase offset ϕ_0, the root mean square of the B0 phase errors for the coils with a good fit were measured to be ϕ_ec= 22.3°±11.8° for the GM dataset, while these values were found to be ϕ_ec= 23.4°±12.3° for the SP trajectory.
Figures 3 and 4 show 4D flow magnitude images of a single slice in one cardiac frame for the GM and SP trajectory, respectively. Both figures show images of the flexible tube without and with eddy current artifact correction, the latter of which were deemed sharper. Both before and after gradient delay compensation and B0 phase error correction, the GM dataset produced visually sharper images than the SP dataset, indicating the value of spatiotemporally uniform k-space coverage for motion and undersampling robustness. The GM and SP trajectory showed comparable phase errors in the calibration data, suggesting that the B0 phase errors affected both images similarly. Additionally, visual image quality improved for both sequences after applying the corrections that were calculated using the calibration scans. The use of a pulsatile flow phantom with cardiorespiratory motion allows for investigation of 3D radial trajectories for 4D flow MRI and the effects of eddy current artifact correction. Further exploration into more radial sampling schemes could be beneficial for future clinical implementation.
Luc DE RUITER (Amsterdam, The Netherlands), Pim VAN OOIJ, Eric SCHRAUBEN
15:40 - 17:10
#47949 - PG444 Initial experiences with monitoring 3D multi-echo gradient echo sequences with a dynamic field camera.
PG444 Initial experiences with monitoring 3D multi-echo gradient echo sequences with a dynamic field camera.
Multi-echo gradient echo MRI sequences are used to acquire images which encode information about tissue properties such as susceptibility and T2* relaxation rate [1]. Measurement of these properties is of great interest in multiple-sclerosis, Alzheimer’s disease, and Parkinson’s disease, where they are promising candidates for quantitative biomarkers of disease progression. Significant progress has been made in quantitative susceptibility mapping (QSM), however application and validation of the technique in vivo remain a challenge [2-4]. Efforts to close the gap between QSM reconstruction algorithm performance in simulation and in vivo have primarily focused on furthering the understanding of microstructural and non-dipolar contributions to the local magnetic field to more completely describe the field-to-source inverse problem at the core of QSM. In contrast, limited work has been done to improve the fidelity of the phase of multi-echo gradient echo imaging data and to understand its influence on QSM accuracy. The use of an expanded encoding model and dynamic field monitoring system may provide an avenue for improving the fidelity of phase data in multi-echo gradient echo imaging [5]. This work describes an initial measurement of higher-order field dynamics during the echo train of a 5-echo multi-echo gradient echo sequence, for both monopolar and bipolar readout strategies.
A template 3D multi-echo gradient echo (meGRE) sequence was developed on a 3 T Philips MR7700 MRI system with the NG2250 XP gradient amplifier (Philips Koninklijke). The 3D non-flyback meGRE scan parameters were as follows: TR = 19 ms, TE1 = 3.4 ms, echo spacing = 1.8 ms, 1x1x4 mm resolution, FOV 230x183x124 mm. A 3D meGRE with flyback (TR = 25 ms, echo spacing = 3.5 ms), and a 2D meGRE without flyback (TR = 86 ms, TE1 = 6.5 ms, echo spacing = 1.7 ms) were also measured in the same session.All scans used a maximum gradient amplitude of 20 mT/m. A Skope dynamic field camera (DFC) was used to monitor a subset of the scan dynamics with a dynamic TR of 280 ms. The scanner software was modified using the Philips pulse programming environment (PPE) to implement changes in sequence timing required by the DFC, and synchronization triggers were added. A calibration sequence was implemented using the PPE. Data from the DFC was recorded using the Skope acquisition system and processed in Skope-fx. Further processing and filtering was performed in MATLAB. Higher order field coefficients were described using maximal phase excursion over a 10cm volume. First-order trajectory variations and resonance frequency drift were assessed, along with higher order field terms. Measurement of spatiotemporal field dynamics for both 2D and 3D Cartesian meGRE acquisitions with and without flyback were demonstrated, validating our implementation of pulse sequence timing changes (see Figure 1). Drift in sampling position compared to the nominal sampling pattern was observed in the first order solid harmonic terms (Figure 2) of up to 15 rad/m. A consistent linear trend was seen in the zeroth order field term throughout the acquisitions, with transient behaviour in the first few seconds of measurement before stabilizing. The transition between transient and linear regimes could be modeled with a simple exponentially modulated linear fit ((1 - e^(-t/T))*(m*t + b), where T describes the time constant of the transient behaviour and m describes the drift in the B0 field with time (Figure 3). Field drifts of between 0.07 and 0.25 Hz per second were measured in the linear regime. The presented work describes our initial experiences with measuring Cartesian meGRE sequences using a dynamic field monitoring system. Separation of the 3rd dimension phase encode gradient from the slab and slice selective rephasing gradients was accomplished without degrading sequence or image fidelity. Observed imperfections in the field dynamics were consistent in magnitude with those reported for echo-planar imaging, as expected. While the observations were consistent with previous work, this measurement is preliminary. Next steps within this work will comprise refinement of the measurement protocol, including optimization of the sequence timings and further investigation of the observed resonance frequency drift. A modified version of a 3D multi-echo gradient echo sequence was developed on the Philips platform which was compatible with dynamic field monitoring, and initial measurement and characterization of system imperfections specific to multi-echo sequences was conducted.
Alexander JAFFRAY (Vancouver, Canada), Julian KLOIBER, Alexander RAUSCHER
15:40 - 17:10
#46513 - PG445 Exact motion simulation vs. retrospective application on static data.
PG445 Exact motion simulation vs. retrospective application on static data.
Calculating the effects of motion in MRI acquisitions is essential to develop more robust sequences, understand the resulting artifacts, and to generate training data for machine learning algorithms for their suppression. This is commonly done by retrospectively distorting the acquired image according to the replicated motion path. We suggest that this approach is physically implausible and can show large differences compared to an accurate simulation. This is because it ignores the complex magnetization dynamics during acquisition, which can influence motion artifacts.
An analytical, physically correct simulation based on Phase Distribution Graphs[1] was used as ground-truth. This simulation was recently extended by the capability of simulating motion[2]. Arbitrary movements during any MRI sequence can be simulated by deriving a closed-form expression for the motion induced phase shift of the magnetization. This phase is produced by the interaction between movement and a variable magnetic field gradient:
ϕ(r_0,T) = ∫_0^T [r(r_0,t) ⋅ g(t) dt]
where ϕ is the phase of a point in magnetization that started at position r_0, accumulated over the time period T, following the trajectory r(r_0,t) through a varying magnetic field with gradient g(t). By segmenting both the movement and the changing gradient field into linearized steps, a closed-form solution can be found:
ϕ = r_0 k_0+1/2 r_0 Δk+1/2 Δrk_0+1/3 ΔrΔk
In the simulation, this phase is calculated per-voxel for the provided motion paths. Furthermore, the phase of magnetization also reacts to refocusing through RF pulses, to their phase, as well as to B_0 inhomogeneities. While the simulation calculates the magnetization based on flip angles, relaxation times and more, the retrospective method works solely on a single image[3]:
y=∑_(t=1)^T [M_t F U_t x]
where the distorted image y is calculated as the sum of all sampled time points. For each point, M_t is the position of the k-space sample, F the Fourier transform, U_t the motion and x the undistorted image.
Both approaches for calculating motion artifacts are compared for two MRI sequences, a TSE acquisition and a balanced SSFP measurement. The TSE sequence uses 120° refocusing pulses, gradient spoiling and has a repetition time of 5 ms. The bSSFP sequence uses 50° pulses with alternating phase and an α\/2 preparation pulse at a repetition time of 10 ms. Both are 2D sequences with a resolution of 64×64, the applied motion is a constant movement in horizontal direction of 5 cm over the duration of the acquisition. Figure 1 displays the comparison of both approaches for the bSSFP sequence. The retrospective method applies motion artifacts directly to the first reconstruction, which stems from a simulation without motion. The result is very close to a full simulation that includes motion, as the difference images show.
Figure 2 shows a similar comparison, this time for the TSE sequence. Here, large differences between the simplistic estimation and a full simulation can be seen. The simulation shows ghosting artifacts which are especially visible in the phase of the reconstructed but are completely missing from the retrospective method. The magnitude difference shows large deviations between both approaches, which are up to 30% of the maximum magnitude of the reconstructed images. While the often-used retrospective approach to generating motion artifacts can indeed capture their overall shape, it can deviate strongly from a full simulation. The difference is stronger for a spoiled sequence. In balanced sequences, the overall phase of the magnetization is set to zero and the motion induced phase shift does not accumulate as strongly. In the TSE sequence however, the interaction between motion and refocusing pulses must be considered for an accurate description of motion artifacts. This is only done in a simulation but not in a retrospective method which only considers an acquired image but not the magnetization dynamics during acquisition. Similar differences could be expected for sequences with strong transient state effects, where the contrast changes during acquisition. Here, the retrospective method could fail in similar ways as this change is missing from the measured signal and requires a simulation to be considered in motion artifact calculations. We propose that the often-used retrospective approach of calculating motion artifacts is insufficient to describe these effects correctly. Machine learning or development of robust sequences could lead to subpar results if the underlying mechanics are not described correctly, as is evident by the partly large differences in reconstructed images. Full simulations should be used if possible.
Jonathan ENDRES (Erlangen, Germany), Moritz ZAISS, Simon WEINMÜLLER
15:40 - 17:10
#47650 - PG446 Recycling field maps: B0 distortion correction using shim calibration scans.
PG446 Recycling field maps: B0 distortion correction using shim calibration scans.
Echo-planar imaging (EPI) [1] is the most common readout strategy for fast MR imaging. It is used in applications such as Diffusion weighted imaging (DWI), BOLD fMRI and ASL. The advantages that it provides in terms of speed come along with an increased sensitivity to field inhomogeneities, arising primarily from susceptibility differences between tissues, which result in image distortions. This effect is stronger for higher background fields and longer readout trains, e.g. in single-shot EPI.
Shimming is a widespread means of mitigating such distortions by counteracting field inhomogeneities through the use of additional hardware. It requires a calibration measurement at the beginning of the scan session. For an accurate shimming procedure, this comprises a field map measurement in the region of interest. Spherical harmonic basis functions are fitted to the field distribution, which can then be realized by actuating the respective shim coils.
Residual distortions remain due to limited capabilities of the shim hardware. These can be addressed by incorporating additional information on the expected distortions in the image reconstruction. Various strategies have been deployed to this end [2–5], among which the most basic method is to acquire another field map, from which a displacement map can be computed, which is then used to unwarp the EPI [6].
In the described imaging procedure, two field maps must be acquired, one for the initial shimming and another one (with modified shim conditions) for the retrospective distortion correction. In this work, we perform both corrections using only one map. This is done by calculating the effect that the difference in shim values has on the field distribution and thereby estimating a new map under the modified shim condition.
Theory:
The field distribution (F) produced by a certain shim state (current vector s of length N_c = #shim coils) at pixel position x can be expressed as shown in Equation 1 (Figure 1), where k counts through the N_sh spherical harmonic basis functions y_k(x). To convert a field map B_A(x) that has been measured under an initial shim state A, to another shim state C, the difference in field distribution must be added to it (Equation 2 in Figure 1). The converted field map is used to compute a displacement map. The un-warping of the EPI images is performed using linear interpolation to the thus distorted coordinates [6].
Experiments:
Experiments were performed on a preclinical 7T small animal scanner (Bruker BioSpec 30/70, Bruker BioSpin GmbH & Co. KG, Ettlingen, Germany), using a transmit volume coil and a 2x2 surface receive array. Measurements were performed using a cylindrical water phantom (MRI PHAN 1H IM M.HEAD, model no. 1P T9660, same vendor) and a naturally deceased mouse.
Field maps were computed from 3D double-echo spin-warp scans. 2D multi-slice single-shot Echo-planar imaging (EPI) was performed using the parameters displayed in the table in Figure 2.
Initial shimming was performed using an FID-based method which only actuates 1st order shims. Subsequently, a field map was acquired and used to optimize the field homogeneity including higher-order shims (here, up to 2nd order and Z^3). In this new shim state, another field map and the EPIs were acquired.
T2 weighted RARE [7] scans were acquired as geometric reference. See Figures 3 and 4. The "recycling" of field maps for B0 distortion correction, presented here, proves to be an effective and timesaving means of image enhancement. There are three potential limitations to the efficacy of the method:
1) The reliability of the method depends on the accuracy of the shim field approximation with a given set of basis functions (here spherical harmonics). If this approximation does not hold, for example, if there were non-linearities with respect to the shim currents that were disregarded in the model, the converted field map would not be valid. However, this behaviour could not be observed in the current study.
2) Since typical shim coils can only be used to produce spatially slowly varying field distributions, usually low-resolution field maps are used for their calibration. However high-resolution field maps might be beneficial for accurate distortion correction. In applications where multiple regions of interest are studied, using only one initial shim calibration, the workflow proposed here might not be the best option, but instead acquiring one low-resolution field map in the beginning and region-specific high-resolution maps for distortion correction so that the proposed conversion would not be strictly necessary (e.g. in Figure 4, if the eyes were the region of interest).
3) In regions where strong field changes lead to dephasing in the spin-warp scan, no reliable field map information can be obtained. In this situation, acquiring a second map under the new shim conditions, where the strong field changes are mitigated, might be conducive to the distortion correction.
Maria ENGEL (Ettlingen, Germany), Martin HAAS, Michael HERBST, Sascha KÖHLER, Christian MEIER, Markus WICK
15:40 - 17:10
#47568 - PG447 Field Inhomogeneity Correction for 2D Single-Shot Lissajous Trajectories.
PG447 Field Inhomogeneity Correction for 2D Single-Shot Lissajous Trajectories.
Single-shot 2D Lissajous trajectories enable full k-space sampling using two widely spaced echo times within a single RF excitation [1]. Fast regridding can be achieved with the Uniform Resampling (URS) algorithm [2], which employs only two matrix multiplications using matrices similar in size to the reconstructed image. However, as the readouts for both echoes are spread across the entire acquisition window, as illustrated in Figure 1, these trajectories are susceptible to off-resonance artefacts, such as blurring and geometric distortion. Fortunately, rapid regridding enables the application of off-resonance correction methods, as described in [3].
In this work, we implement the time-segmented off-resonance correction from [3] and its iterative counterpart [4] for the reconstruction of 2D Lissajous trajectories and compare them using simulations, phantom and in vivo measurements.
To evaluate the performance of the off-resonance correction, a fully sampled, single-shot 2D Lissajous sequence (0.24 cm² FOV, matrix size: 96x96, TE: [8.7ms, 108.9ms]) was implemented using PULSEQ [5] The total readout duration was 113.6 ms, including two navigator lines at the beginning for gradient delay correction. The readout started 4ms after RF-excitation.
Simulations were performed with MRZero [6]. Phantom and in vivo measurements were acquired on a 3T Siemens Magnetom Prisma system. B₀ field maps and coil sensitivity profiles were estimated using a dual-echo GRE acquisition with echo times of 4.92 ms and 7.38 ms. The B0 field maps were calculated from the phase differences of the two echoes. Coil sensitivity maps were estimated using the ESPIRiT algorithm from the BART toolbox [7, 8]. Thirty time segments were used to reconstruct the simulations. The simulated field map ranged from –11 Hz to 45 Hz. As shown in Figure 2, the uncorrected images exhibit distortions and blurring (indicated by red arrows). The off-resonance correction eliminates most of the artefacts. However, residual artefacts are still visible in areas with strong field fluctuations. Overall, the CG-SENSE reconstruction with off-resonance correction provided sharper images with less blurring than the faster URS reconstruction for both echoes. Testing up to 100 time segments did not result in a reconstruction with fewer residual artefacts.
In the phantom measurements, 16 time segments were sufficient to correct nearly all distortions. Nevertheless, some off-resonance artefacts remained in the second echo. The corresponding field map ranged from –19 Hz to 14 Hz.
In in vivo data, neither the time-segmented URS nor the CG-SENSE-based method could fully correct for signal dropout or strong distortions due to B₀ inhomogeneity (field map range: –18 Hz to 150 Hz). Still, both correction methods reduced image blurring and improved grey-to-white matter contrast. The images from the reconstruction using 35 time segments are shown in Figure 4. Increasing the number of time segments to 100 did not improve the reconstruction quality. For moderate B₀ inhomogeneities, time-segmented URS and the time-segmented CG-SENSE reconstruction can effectively reduce blurring and distortion in Lissajous measurements. However, for larger field deviations – especially in in vivo measurements – the effectiveness of these corrections is limited. With the current spatial resolution of 2.5 mm, the readout time of 113 ms may be too long for complete correction of such artefacts. Due to the nested acquisition of echo times, 2D Lissajous trajectories are also particularly sensitive to B₀-related effects. Furthermore, intra-voxel dephasing can increase the signal loss for the large voxel size of the sequence used in this work [9]. The time-segmented off-resonance correction methods used in this work can compensate for most artefacts in simulated data and phantom measurements of 2D Lissajous trajectories. However, significant artefacts remain in in vivo images in regions with strong field inhomogeneities. Further improvements could be achieved by combining these corrections with trajectory undersampling and/or multishot methods that reduce the overall readout duration and/or permit smaller voxel sizes.
Felix LANDMEYER (Jülich, Germany), Markus ZIMMERMANN, Fabian KÜPPERS, Seonyeong SHIN, N. Jon SHAH
15:40 - 17:10
#47793 - PG448 End-to-End Pipeline for GIRF Correction of Pulse Sequences in Julia.
PG448 End-to-End Pipeline for GIRF Correction of Pulse Sequences in Julia.
Magnetic resonance imaging (MRI) relies on precise control and knowledge of magnetic field gradients to produce high-quality images. Gradient imperfections can lead to artifacts that compromise image accuracy, particularly in quantitative MRI applications where exact knowledge of the gradients is crucial. Gradient impulse response functions (GIRFs) provide a robust approach to characterizing and correcting these imperfections by modeling the gradient system's dynamic response to input signals [1].
In this work, we introduce a computational pipeline developed in the Julia programming language for computing and applying GIRFs based on magnetic field measurements. This pipeline builds on and extends existing implementations [2] by integrating GIRF corrections directly into MRI sequences stored in Philips GVE files. It offers an efficient alternative to MATLAB-based implementations while maintaining compatibility with the Julia ecosystem. Importantly, our pipeline extends GIRF computation beyond first-order field measurements to include second- and third-order solid harmonic terms, allowing for a more comprehensive correction of gradient-related artifacts.
The computational pipeline was implemented in Julia, chosen for its high performance, intuitive syntax, and modular design. Julia’s support for efficient multi-threading and extensive package library makes it well-suited for computationally intensive tasks.
The pipeline utilizes the GVE.jl module for loading Philips GVE files and extracting the gradient waveforms and sequence timing information [3]. The waveforms are interpolated to match the scanner’s dwell time. GIRFs are computed based on field measurements using frequency-domain division, following the process described by Vannesjo et al. [1]. Alternatively, externally provided GIRFs can be imported. The GIRF correction step involves transforming the input gradients into the frequency domain, followed by a multiplication with the GIRF in frequency domain and subsequently performing an inverse Fourier transform to obtain the corrected gradient waveforms in the time domain. To maintain precision, both the input gradients and the GIRF are interpolated in the frequency domain to match the bandwidth of the gradient system.
To demonstrate the usage of our proposed pipeline, the GIRF was computed from 12 triangular gradient waveforms with varying strengths measured on a Philips MR7700 3T system with the NG2250 XP gradient system. Magnetic field measurements were acquired using a dynamic field camera consisting of 16 spatially distributed NMR probes, enabling the representation of the magnetic field as a solid harmonic expansion up to third order [4]. These high-order terms allow the pipeline to correct for spatially varying gradient imperfections. The Julia-based pipeline efficiently loaded and processed Philips GVE files and applied GIRF corrections to gradient waveforms with minimal computational effort. Corrected gradients exhibited temporal evolution consistent with the low-pass filtering effects of the GIRF.
For evaluation, GIRF corrections were applied to a diffusion-weighted imaging sequence. Figure 1 shows the computed first-order GIRF and demonstrates the low-pass behaviour. Figure 2 illustrates the corrections in gradient waveforms for a selected time interval within the sequence. The computational efficiency of the pipeline underscores Julia’s suitability for high-performance MRI simulations and reconstruction tasks. The proposed Julia-based pipeline is an advancement in integrating GIRF corrections into MRI simulation and reconstruction projects. Its direct compatibility with Philips GVE files eliminates the need for intermediate file conversions, streamlining data processing and reducing potential sources of error. This direct integration not only simplifies the pipeline but also ensures the fidelity of the input data. The inclusion of higher-order solid harmonic terms extends the GIRF framework, allowing for precise correction of spatially complex gradient imperfections. Julia's modular architecture ensures that the pipeline can be easily extended to include additional functionalities, such as integration with MRIReco.jl for reconstruction workflows or KomaMRI.jl and others for simulation [5-6]. We developed an efficient Julia-based pipeline for computing and applying GIRF corrections to Philips GVE MRI sequences. By incorporating gradient field measurements up to third-order solid harmonics, the pipeline streamlines characterization of gradient imperfections and enables precise corrections. This improves the fidelity of gradient waveforms, leading to higher-quality image reconstructions.
Julian KLOIBER (Vancouver, Canada), Alexander JAFFRAY, Alexander RAUSCHER
15:40 - 17:10
#47936 - PG449 Tissue Boundary Artifact Reduction Performances of Helmholtz-MREPT and cr-MREPT.
PG449 Tissue Boundary Artifact Reduction Performances of Helmholtz-MREPT and cr-MREPT.
Magnetic Resonance Electrical Property Tomography (MREPT) is a non-invasive reconstruction technique for imaging tissue electrical properties from MRI data. In this study, two widely used approaches, the Helmholtz-equation based (Helmholtz-MREPT) and the convection–reaction-equation based (cr-MREPT) methods, are compared in terms of artifacts at tissue boundaries, using both simulated and experimental phantoms.
In Helmholtz-MREPT approach, based on the local homogeneity assumption, conductivity is computed, pixel-wise using the equation in Figure 1.a.
Direct calculation of the phase Laplacian is extremely noise‐sensitive. Thus, a local weighted parabolic‐curve fit is applied within a 3D kernel. These weights are computed according to a Gaussian function of the MR magnitude difference between each kernel point and the local center as shown in Figure 1.b. The standard deviation, s, is taken as 0.45.
Following reconstruction of the conductivity maps, a weighted median filter is applied to further enhance the performance near boundaries.
The second method, cr‐MREPT, solves a convection–reaction PDE simultaneously across the entire grid, without relying on MR magnitude or local homogeneity assumptions. The convection-reaction PDE equation is shown in Figure 1.c.
The equation is discretized on either a Cartesian grid or a triangular mesh via the finite‐element method and solved for resistivity. Conductivity is then derived as the reciprocal of the computed resistivity. A small diffusion parameter (c=0.005) regularizes the solution to prevent abrupt changes.
The described algorithms are evaluated on both digital and physical (real) phantoms. Three digital phantoms with different conductivities were simulated in COMSOL at 128 MHz. Phantom-1 has four anomalies with conductivities 0.75–1.25 S/m against a 0.5 S/m background, whereas Phantom-2 and Phantom-3 have small anomalies with 1 S/m conductivity with the same background. Since boundary artifacts obscure especially small repetitive objects, Phantom-3 is generated which contains much smaller anomalies compared to Phantom-2, with diameters of 0.3–0.6 cm. Both the noise-free and noisy versions of Phantom-3 were evaluated. Phantom-4, an experimental agar‐gel (20 g/L Agar, 2 g/L NaCl, 0.2 g/L CuSO4) phantom with 4.5 mm and 7 mm salt‐water anomalies (6 g/L NaCl, 0.2 g/L CuSO4) are scanned at 3 T using a bSSFP sequence (FA=40°, TE/TR=2.35/4.7 ms, 1.56 mm isotropic, NEX=32) at Bilkent University UMRAM. All algorithms are implemented in MATLAB. In Helmholtz-MREPT reconstructions, boundary artifacts are nearly eliminated around the anomalies larger than 1 cm in Phantom-1 and Phantom-2, and the reconstructed conductivity values closely match the true values. However, experiments with Phantom 3, which consist of smaller anomalies, demonstrate that although anomalies with 0.3 cm diameter can still be identified in noise-free setting, noise addition yields severe errors in anomaly boundaries and reconstructed conductivity around transition regions. While the weighted median filter, especially in the noisy setting, mitigates some artifacts and modestly refines conductivity values, it does not fully alleviate the problem. The same pattern is also observed in the physical phantom (Phantom-4) scan: anomaly edges remain sharp, but reconstructed conductivity near those transitions continues to exhibit errors. The provided profile plots (along the central line of the middle slice) for each phantom clearly illustrates these results.
Regarding the conductivity maps reconstructed via cr-MREPT method, similar to the Helmholtz-MREPT method, boundary artifacts are nearly eliminated around large anomalies in Phantom-1 and Phantom-2 with near-true conductivities. However, smaller anomalies exhibit underestimated conductivities, especially in Phantom-3. Adding noise to Phantom-3 does not introduce significant boundary artifacts in reconstruction but produces a smoother conductivity distribution. These findings are confirmed also in the physical experiments conducted with Phantom-4. The corresponding profiles further highlight these trends. Conductivity distributions reconstructed by Helmholtz‐MREPT and cr‐MREPT, are compared in terms of artifacts at tissue boundaries, using both simulated and experimental phantoms. Overall, both algorithms demonstrate successful performance but exhibit distinct strengths. The Helmholtz-MREPT approach detects anomaly boundaries sharply but can introduce errors in conductivity estimates and artifacts near edges. By contrast, the cr-MREPT method offers greater robustness against noise. However, its low-pass filtering effect tends to smooth the conductivity distribution and softens boundary definition.
Elif YALI (Ankara, Turkey), Emine Ulku SARITAS, Yusuf Ziya IDER
15:40 - 17:10
#46693 - PG450 Optimizing Echo Planar Imaging for Improved BOLD Sensitivity in Deep Gray Matter Structures.
PG450 Optimizing Echo Planar Imaging for Improved BOLD Sensitivity in Deep Gray Matter Structures.
fMRI typically uses echo-planar imaging (EPI), which is highly vulnerable to field inhomogeneities causing signal dropout and geometric distortions. Optimizing EPI parameters, such as phase encoding (PE) direction, tilting slices to distribute susceptibility-induced field gradients across multiple axes, and incorporating z-shimming offers a valuable tool for maximizing Blood Oxygen Level Dependent (BOLD) Sensitivity (BS) in targeted brain regions.
Previous studies have estimated BS gains by specifying a global effective transverse relaxation rate (R₂*) [1,2]. However, this neglects anatomical variation within and between ROIs. In this study, we investigated the impact of incorporating spatially varying R₂* values on BS optimization, focusing on iron rich deep gray matter structures—the putamen and pallidum—which have distinctly high R₂* values. These structures play a crucial role in many cognitive functions and are affected in a range of neurological disorders. The hippocampus was also evaluated for comparison.
B0 field maps and high resolution (1mm³) R₂* maps from a large cohort (n=138; Fig. 1) spanning a broad age range were used [3] to model the impact of susceptibility-driven magnetic field gradients. BS was estimated using analytical expressions [2,4], considering a standard 3T 2D EPI protocol (transverse orientation, TE = 30 ms, echo spacing = 0.5 ms, resolution = 3×3×3 mm³). Optimization was performed by systematically varying PE direction (posterior-anterior, PA vs. anterior-posterior, AP), slice tilt (-45° to 45°), and z-shim gradient moment (-5 to 5 mT/m*ms).
Regions of interest—putamen, pallidum, and hippocampus—were defined using the AAL3 atlas [5]. The field gradients in the PE and slice-selective directions were quantified within these ROIs. Protocol optimization was carried out either (1) using a global R₂* (1/45 ms⁻¹) or (2) using voxel-wise R₂* values. Analyses were performed separately for left and right hemispheres to assess potential asymmetries. Average PE field gradients were -2μT/m, 10μT/m, 21μT/m in the hippocampus, putamen and pallidum, respectively. In the slice direction, these were 25μT/m, 4.8μT/m and 6μT/m.
Optimal EPI parameter sets that yielded the highest mean BS varied by ROI but were largely consistent across hemispheres (Fig. 2). The putamen and pallidum specifically required AP phase-encoding with moderate slice tilts (20° and 10°) to optimally mitigate BS loss. The gains over an optimal PA protocol were substantial (18.9% and 8.3% for the pallidum and putamen respectively, c.f. Fig. 3). For the hippocampus, optimal settings aligned with previous theoretical and empirical studies [1,2]. Opposite PE direction and tilt were obtained for the left and right hemispheres—though these are close to degenerate in the hippocampus (c.f. Fig 3).
The optimal parameter sets were largely independent of how R₂* was incorporated into the simulations though a voxel-wise R₂* marginally increased the predicted BS in the hippocampus (Fig. 4). Field gradient maps alone cannot account for microstructural susceptibility variations. Relying solely on a global R₂* value limits model accuracy. Our use of a spatially-resolved R₂* map from a large cohort improves anatomical fidelity without compromising reliability.
The hippocampus with a relatively uniform and small PE gradient profiles was most impacted by voxel-wise R₂* modelling. In contrast, the putamen and pallidum experience much stronger local PE gradients, potentially causing macroscopic field inhomogeneities to dominate over R₂* variability.
The putamen’s average R₂* (21.9 s⁻¹) closely matched the global value (22.2 s⁻¹) used in the simulations which minimized the impact of voxel-wise modelling. The hippocampus exhibited considerably more spatial uniformity in R₂*, but with a much lower mean of 15.7 s⁻¹. This increased the predicted BS that could be achieved, though the optimal protocol settings were unaffected. The pallidum, while having a higher average R₂* (33.6 s⁻¹) and greater spatial variance yielded similar optimal settings whether a global or voxel-wise value was used (Figs 2,3).
These simulations only considered transverse orientations and used a fixed TE, limiting the potential strategies available to overcome inherent susceptibility gradients. Optimizing for such diverse macroscopic and microscopic field gradients would benefit from spanning a broader range of settings, and will be the focus of future work. Under the conditions investigated, our findings suggest that the impact of R₂* variability is secondary to macroscopic field homogeneity effects and therefore has limited impact on the optimal protocol settings.
Cognitive neuroscience studies typically involve multiple ROIs, where protocol design should consider incorporating knowledge of the underlying microstructure and adopting a constrained optimization approach for balancing trade-offs between ROIs.
Shokoufeh GOLSHANI (London, United Kingdom), Martina CALLAGHAN
15:40 - 17:10
#47505 - PG451 Improving the motion robustness of breath-holding in abdominal 3D T1 gradient echo imaging using a Cartesian acquisition with spiral profile ordering.
PG451 Improving the motion robustness of breath-holding in abdominal 3D T1 gradient echo imaging using a Cartesian acquisition with spiral profile ordering.
Abdominal imaging typically involves multiple breath-hold acquisitions to mitigate respiratory motion artifacts. However, patients' breath-hold capabilities may not cover the full scan duration, leading to incomplete (early respiration onset) or late breath-holds (delayed breath-hold start). When motion artifacts occur, the breath-hold usually must be repeated. Acquiring motion-free central k-space data is crucial to reduce artifacts [1].
Clinically, 3D Cartesian trajectories with pseudo-random undersampling via Compressed Sensing are used in gradient echo imaging to reduce scan times [2], and breath-holds are performed at end-expiration to minimize artifacts [3].
Recently, Cartesian Acquisition with Spiral Profile Ordering (CASPR) has been proposed for free-breathing abdominal imaging [4-7], offering central k-space oversampling that enhances motion robustness and enables self-gating without external navigators [8]. Free-breathing self-gating typically acquires 1D profiles in the feet-head (FH) direction, the most affected by respiratory motion [8], but FH frequency encoding reduces efficiency for high resolution 3D axial abdominal imaging, prolonging scan and breath-hold durations. To improve motion robustness, this work proposes a method using a CASPR trajectory with anterior-posterior (AP) frequency encoding and self-gating to correct respiratory motion artifacts in incomplete and late breath-hold 3D axial liver MRI.
Five volunteers were scanned at 3T (Ingenia Elition X, Philips) using a T1-weighted 3D gradient echo sequence with AP frequency encoding (TE=1.32/2.42ms, TR=3.73ms, scan time=15.8s, voxel size=1.47x1.59x5mm, FOV=358x537x265mm) and a spiral-in-spiral-out golden-step CASPR trajectory (30 profiles/shot, 112 shots). For one subject, a conventional Cartesian acquisition without k-space center oversampling and with pseudo-random undersampling (acceleration factor=7.55) was acquired to compare with the CASPR scan. Each volunteer was asked to perform a perfect, an incomplete breath-hold at end-expiration, and a late breath-hold.
Self-gating was performed based on the oversampled central k-space line in AP direction. Coils with sufficient AP motion were selected via coil-wise frequency analysis (low amplitudes for frequencies >1Hz) of the first principal components from the oversampled k-space line. Principal component analysis was conducted on the coil signals to determine a respiratory motion curve.
Breath-hold periods were identified using a second-order central finite differences method: curve segments were labelled as breath-holds if the derivative of the curve's moving average (window=7) stayed close to zero over a continuous interval lasting at least 20% of the scan duration.
Three reconstruction strategies were explored for the CASPR data. First, images were reconstructed without retrospective motion correction (Fig. 1.1). Then, soft gating and hard gating approaches for motion correction were investigated. For both, data acquired in the breath-hold phase, detected by the previous algorithm, was assigned a weight of one. Soft gating applied a Gaussian weighting centered on the average amplitude of the breath-hold segment to the non-breath-hold data (Fig. 1.2), while hard gating rejected it entirely (Fig. 1.3). All cases were then reconstructed using an iterative reconstruction algorithm with spatial total variation regularization. Fig. 2 compares a traditional Cartesian acquisition without k-space center oversampling to CASPR for perfect, incomplete and late breath-holds, showing comparable reconstruction results without gating. Motion artifacts are visible for incomplete and late breath-holds also for the CASPR acquisition. Soft and hard gating in the CASPR scans can significantly improve the image quality, with both gating methods yielding similar results during breath-holds at end expiration (Fig. 3). When the breath-hold is in a state different from the end-expiration state, soft gating showed reduced performance in the incomplete breath-hold scan (Fig. 4). This work proposes a CASPR acquisition for liver breath-hold 3D gradient-echo imaging and compares two respiratory self-gating approaches for motion artifacts correction. The method improves image quality for incomplete and late breath-holds. The inherent self-navigator in the AP direction allows motion estimation even though it is less affected by motion than the FH direction. Current results do not show a clear improvement for soft-gating compared to hard-gating. While the proposed method can improve image quality in motion-affected examinations, scans may remain non-diagnostic if too little k-space data unaffected by motion is available. A novel breath-hold acquisition strategy using a CASPR trajectory with a self-navigator signal in the AP direction was proposed. Results demonstrate the ability of the self-navigated CASPR to improve the motion robustness of 3D liver gradient echo imaging in incomplete and late breath-hold scenarios.
Alice SCUDELETTI (Munich, Germany), Jonathan STELTER, Kilian WEISS, Rickmer BRAREN, Dimitrios C. KARAMPINOS
15:40 - 17:10
#45859 - PG452 MORSE-PI: Refinement and validation of flexible and robust structural and functional phase imaging.
PG452 MORSE-PI: Refinement and validation of flexible and robust structural and functional phase imaging.
We recently proposed MORSE-PI [1]: a robust and computationally efficient method for coil sensitivity estimation and complex (magnitude and phase) image reconstruction using a regularised SENSE [2] formalism. MORSE-PI produces high-quality images, free of aliasing artefacts and phase singularities, for both structural and functional data, enabling simultaneous magnitude- and phase-based functional or quantitative MRI including high quality Quantitative Susceptibility Mapping (QSM).
MORSE-CODE, the precursor reconstruction framework, involves voxel-wise singular value decomposition with an intrinsic singular vector sign and phase ambiguity, often yielding phase singularities, which limits options for QSM calculation and yields sub-optimal results. To overcome this limitation, MORSE-PI computes a Virtual Reference Coil (VRC) [3] to correct the phase of the estimated coil sensitivities. Here, we improve the robustness of the VRC calculation and subsequently evaluate MORSE-PI by comparing image quality against other state-of-the-art image reconstruction methods, and quantifying reproducibility.
MORSE-PI uses k-space calibration data, embedded within the accelerated acquisition or acquired separately, to estimate complex coil sensitivities. Prewhitening, using a measured noise covariance matrix, improves image SNR by decorrelating the coil channels. However, for VRC creation in MORSE-PI we instead increase the cross-channel correlation to achieve a robust VRC with signal support over the entire field of view. This is achieved by repeatedly applying the inverse of the prewhitening matrix. Doing so twice has proved sufficient to create singularity-free phase results in diverse scenarios including EPI and gradient echo (GRE) readouts, at 3T and 7T with 64 and 32 channel head coils respectively.
The final VRC is a voxel-wise complex sum of coil sensitivities, after scalar phase matching. The reference voxel was originally [1] the image centre. However, using the phase value from the weighted centroid of the image improves robustness to specific head position within FOV. The VRC phase is subsequently used to correct the phase of the original prewhitened MORSE-PI coil sensitivities. The final magnitude and singularity-free phase images are reconstructed using a regularized SENSE formalism. The method is deployed as a MATLAB-based gadget within Gadgetron [4] for real-time reconstruction. MORSE-PI has been deployed for a diverse range of 3T and 7T studies in our department, yielding high quality magnitude and singularity-free phase images. Protocols used to illustrate exemplar results are listed in Fig 1.
Fig.2 compares 3D GRE MORSE-PI results at 7T with three alternative image reconstruction methods tailored to phase imaging. GRAPPA+Adaptive-Combined and ESPIRiT are corrupted by phase singularities, yielding image artefacts (red arrows), whereas GRAPPA+ASPIRE and MORSE-PI both provide singularity-free phase images and high-quality CLEAR-SWI and QSM results.
Fig.3 is a quantitative comparison of scan-rescan QSM based on 3D GRE acquisitions reconstructed with GRAPPA+ASPIRE and MORSE-PI at 3T and 7T. The histograms show higher standard deviation for GRAPPA+ASPIRE than for MORSE-PI, across field strengths and contrasts, but particularly at 3T.
MORSE-PI can be applied flexibly whereas GRAPPA+ASPIRE requires multi-echo data. Fig.4 evaluates the temporal stability of MORSE-PI based magnitude and QSM results derived from single echo, slab-selective high-resolution 3D EPI [5] over 50 volumes. Various QSM processing pipelines can be used, including NORDIC denoising [6,7], showcasing MORSE-PI’s flexibility. MORSE-PI calculates coil sensitivities and corrects their phase using a VRC approach. The original implementation [3] calculated the VRC from coil-wise reconstructed complex images, rather than sensitivities, which can lead to focal regions without VRC support and therefore phase singularities [8]. We ensure here that the VRC estimate is supported over the entire volume of interest by (1) estimating the VRC sensitivity in a heavily correlated coil space, rather than in the prewhitened space in which the sensitivities are in general steeper and more likely to yield poor VRC support, and (2) using a weighted centroid to more robustly select the phase-matching voxel. MORSE-PI performed well against state-of-the art phase imaging approaches with enhanced flexibility for protocol and processing choices (c.f. Fig.4). MORSE-PI achieved good reproducibility (GRE) and temporal stability (EPI) of QSM results. MORSE-PI flexibly provides reproducible, high SNR, fold-over-free and singularity-free phase images, as demonstrated for single-echo and multi-echo structural GRE and functional EPI scans. MORSE-PI naturally lends itself to imaging techniques, such as structural and functional QSM, that necessitate high-quality phase images without additional scan time or compromising magnitude image quality.
Barbara DYMERSKA (London, United Kingdom), Oliver JOSEPHS, Nadine GRAEDEL, Vahid MALEKIAN, Callaghan MARTINA
15:40 - 17:10
#47771 - PG453 Partial Volume Effects across tumor regions in ASL perfusion imaging of Glioblastoma.
PG453 Partial Volume Effects across tumor regions in ASL perfusion imaging of Glioblastoma.
Glioblastomas (GBM) are malignant brain tumors with a 2-year survival rate of ~27% [1]. MRI is the preferred technique for diagnosis and monitoring of GBM. Perfusion MRI helps assess treatment response and possible recurrence, as increased perfusion suggests angiogenesis associated with tumor growth [2]. Arterial Spin Labelling (ASL) stands out for delivering cerebral blood flow (CBF) maps without the need for exogenous contrast agents [2]. Unfortunately, it suffers from intrinsically low signal-to-noise ratio and poor spatial resolution [2-3], which can lead to Partial Volume Effects (PVEs). Although Partial Volume (PV) correction methods have been developed for ASL perfusion imaging [3-4], these were aimed at resolving PVEs between gray matter, white matter, and CSF. Here, we aim to assess and correct PVEs across the three tumor regions of interest (ROIs) that are usually considered in GBM assessment: enhancing tumour (ET), necrotic and non-enhancing tumor core (NCR/NET) and edema.
Data from 3 illustrative GBM patients were analysed in this study, including pseudo-continuous ASL (PCASL, TR/TE =91.42, TI=2.025 s, readout= 3D stack of spirals fast spin echo with 6 arms, 2 repetitions, voxel size = 1.875x1.875x4.000mm³) and T1-weighted contrast-enhanced (T1CE) images acquired on a 3T GE system (Fig. 1).
T1CE images were skull-stripped and registered to Brain Tumour Segmentation (BraTS) space with 1mm3 isotropic resolution. Three ROIs (ET, NCR/NET and edema) and a fourth class representing the rest of the brain were defined using an ensemble deep learning model that combines the outputs of two networks: a ResNet and a UNet with a Swin transformer as the encoder [5].
Registration between the T1CE and PCASL images was performed using an affine transformation with the ANTsPy library. The 3 segmented ROIs and rest of the brain class were transformed into the PCASL space and normalized in such a way that the sum of the 4 classes is 1 in each voxel, yielding the PVs for each class: wk, j (for each voxel j and class k). Relative CBF (f) maps were obtained by averaging the control-label images across repetitions. The mean CBF (f) was then calculated for each tumor ROI, defined by considering PV thresholds t between 10% and 100% in 1%-steps (down to a minimum of 10 voxels per ROI).
PV correction was performed by assuming a constant f value in each tumor ROI: fNCR/NET, fET and fedema, and then solving the following equation to estimate them: fvoxel j=k in {NCR/NET, ET, edema}wk, j.fk (2)
First, initial values were obtained by linear regression of f as a function of PV threshold in each ROI. Then, the system was solved iteratively using different loss functions (least squares, soft_l1, and huber), using “least_squares” function from SciPy’s optimization module. f (Fig. 2) increases with threshold in ET, indicating PVEs from surrounding low-perfusion tissue. In edema, f decreases as threshold rises, likely due to exclusion of high-perfusion voxels (namely, ET and NCR/NET). NCR/NET shows patient-specific trends: stable in patient 1, decreasing in patient 3, and inconclusive in patient 2, where voxel count is lowest.
In patient 1, histogram analysis (Fig. 3) confirms these trends: thresholding excludes low-f voxels in ET and high-f voxels in edema, shifting the mean accordingly. In NCR/NET, voxel removal is balanced around the mean, resulting in minimal net change. Similar patterns appear across patients.
Solving equation (2) produced fNCR/NET, fET and fedema estimates (Fig. 2), which, when applied to original CBF maps, yielded corrected maps. Estimates varied across patients and methods, with loss-based approaches showing possible presence of mild outliers, since estimates were lower when compared to least-squares loss. Moreover, least-squares loss provided estimates closer to the initial guesses. This is expected, as the least-squares loss minimizes the sum of squared residuals, naturally favoring solutions closer to the initial estimate (in this case, the mean CBF at t = 100%). Rising f in ET supports higher perfusion relative to surrounding tissue, with PVE driving the trend as low-perfusion contamination is reduced. Decreasing f in edema reflects exclusion of high-perfusion voxels from neighboring regions. NCR/NET behavior is less consistent due to low voxel count and regional overlap, limiting conclusions despite its expected low perfusion.
Variability in the estimated volume fractions (PVs) across patients and methods highlights individual anatomical and segmentation differences, probably affected by tumour sizes. Since PVEs are minimized, corrected maps offer better delineation of perfusion characteristics across tumor subregions. This study highlights the presence of PVEs in ASL CBF maps across the GBM regions, potentially affecting ROI-based analysis, including radiomics approaches. More accurate perfusion estimates in tumor regions could enhance treatment planning and therefore contribute to GBM recurrence reduction.
Afonso SIMÕES (Lisboa, Portugal), Catarina PASSARINHO, Marta P. LOUREIRO, Ana MATOSO, Pedro VILELA, Rita G. NUNES, Patrícia FIGUEIREDO
15:40 - 17:10
#47805 - PG454 Finding the optimal saturation scheme for combined detection of APT and NOE effects. Application to Parkinson's disease at 3 T.
PG454 Finding the optimal saturation scheme for combined detection of APT and NOE effects. Application to Parkinson's disease at 3 T.
Parkinson’s disease is a neurodegenerative disorder characterized by physiological changes in the brain. One hypothesis is the aggregation of the alpha synuclein (α syn) protein, leading to neuronal death [1]. Chemical Exchange Saturation Transfer (CEST) MRI provides insights into protein content through amide proton transfer (APT) contrast [2] and the Nuclear Overhauser Effect (NOE) [3]. Standard CEST imaging requires long saturation pulses, extending scan time and limiting clinical implementation. In addition, CEST signal depends on the pulse sequence features and parameters used, which may lead to differences in image contrast and interpretation. This study aims to identify an optimal saturation scheme to achieve high APT and NOE effects while minimizing acquisition time.
Simulation: To first investigate acquisition parameters on NOE and APT-CEST effects, simulations were performed using a three-pool model, based on and adapted from scripts available in the pulseq-cest-library (https://github.com/kherz/pulseq-cest-library).
Phantom: A phantom with raw and coagulated egg white (REW and CEW) was prepared, as egg white is a suitable model for mobile protein amide protons [4] (Fig. 1).
Healthy volunteers: Four healthy volunteers (HV), from 20 to 50 years old with no history of neurological disorder (Clinical trial NCT05107232; Univ. hospital of Rennes).
MRI data acquisition: All phantoms and HV data were acquired at 3 T (Magnetom Prisma VE11C; Siemens Healthineers; Erlangen; Germany) using a 64-channels head coil. The 3D gradient recalled echo (GRE) CEST snapshot sequence [5] was used. For the saturation scheme, a total of 29 saturations offsets were swept from -6.0 to 6.0 ppm every 0.5 ppm, with repeats at -3.5 and 3.5 ppm (n=3 each). The unsaturated reference was acquired at -300 ppm. Several saturation parameters were investigated to identify the optimal conditions for maximizing the APT ratio such as the number of pulses (np, from 10 to 50 every 10) and the B1 (from 0.6 to 3.0 µT every 0.1 µT). The time pulse tp and delay td were fixed at 50 ms and 5 ms respectively. The GRE readout parameters followed literature standards [5]. For phantoms, acquisitions were repeated five times in different MRI sessions and a Magnetization Prepared Rapid Acquisition Gradient Echoes sequence and a Turbo Spin Echo sequence were used to quantify T1 and T2 values respectively, in REW and CEW.
For HV, an additional 3D T1w (spatial resolution of (1 mm)3) was acquired to segment cerebral regions of interest (ROI).
Processing: CEST images were analyzed using MATLAB and the adapted CEST_EVAL toolbox (https://github.com/cest-sources/CEST_EVAL). Two ROIs were defined in REW and CEW. MTRasym was computed to compare saturation schemes. In HV, gray/white matter and six deep gray nuclei were segmented from T1w images using FreeSurfer [6]. Simulations pre-identified optimal saturation parameters such as np=35. Fig. 2 shows the simulated Z-spectra and MTRasym curves for REW vs CEW, using in vitro quantified T1 and T2 and the previously optimized B1 and np. A clear distinction between REW and CEW is visible, reflecting protein denaturation on the CEST signal.
For phantom CEST acquisitions, the minimum np was 35 for reliable analysis. Fig. 3 shows the APT-ratio maps for varying B1 values, showing increased APT contrast with B1. Full Z-spectra and MTRasym in REW vs CEW in Fig. 4 confirmed these observations, with distinct dips at 3.5 ppm in every Z-spectrum (Fig. 4.A-B), less pronounced in CEW which is consistent with less amide protons exchanging with bulk water. The MTRasym (Fig. 4.C) peaked at 3.5 ppm for every B1. More precisely, the highest ratio appeared at B1=2.2 µT with a MTRasym=0.16. From this peak, the MTRasym values decreased consistently down to 0.04 for B1=0.6 µT. Fig. 4.D showed less intense peaks as expected, due to a reduction of protons exchanges and NOE effect. The highest difference between MTRasym of REW vs CEW at 3.5 ppm was spotted at B1=2.2 µT (Fig. 4.E), demonstrating the optimum B1 value to differentiate both forms of proteins.
Preliminary in vivo data were coherent with our initial in vitro observations as APT CEST signal was higher in gray matter than in white matter due to its greater content of mobile proteins. This study optimized the CEST saturation scheme of the 3D GRE snapshot CEST sequence [5] to achieve high APT and NOE contrast at 3 T. Saturation schemes and parameters tested demonstrated a substantial impact on the resulting Z spectrum and MTRasym. Other parameters can also have an impact such as the duty cycle as well as the pulse shape. Further analysis is ongoing to refine these parameters by simulating a three-pools model to compute the Z-spectrum. For a 3 T acquisition on mobile proteins, we have identified a saturation scheme that maximizes the APT and NOE ratio. Future work will focus on investigating different conformations of α-syn, given its relevance in the context of Parkinson’s disease.
Aurélien HERVOUIN (Rennes), Johanne BEZY-WENDLING, Fanny NOURY
15:40 - 17:10
#46660 - PG455 Physics-Inspired Coil Profile Estimation for Accelerated Phase-Cycled bSSFP Imaging.
PG455 Physics-Inspired Coil Profile Estimation for Accelerated Phase-Cycled bSSFP Imaging.
Balanced Steady-State Free Precession (bSSFP) imaging offers high signal-to-noise ratios (SNR) but is highly sensitive to field inhomogeneities, leading to banding artifacts [1]. Phase-cycling effectively reduces these artifacts by shifting the bandings across images but significantly increases scan time [2]. To overcome this limitation, strategies similar to parallel imaging have been proposed to accelerate across multiple acquisitions. A banding-free image can then be reconstructed by treating phase-cycled images as virtual coils [3, 4].
A key challenge in this approach is the accurate estimation of bSSFP profiles. Although off-resonances vary smoothly, the spectral response of bSSFP causes rapid intensity variations associated with banding artifacts in image space. Conventional methods, which simply use the low-frequency information from auto-calibration signals can thus not fully represent these transitions and suffer from inaccuracies in the estimated profiles. We introduce a physics-inspired approach that leverages off-resonance information directly from auto-calibration signals. Thus, the bSSFP signal behavior is more accurately reflected and reconstruction quality improves, especially at high acceleration factors.
Experiments were conducted on a 1.5T scanner (neo315, Neoscan Solutions GmbH) using a multi-compartment phantom with five tubes containing different PVP concentrations [5]. Images were acquired using a 3D phase-cycled bSSFP sequence (flip angle = 50°, TR/TE = 6/3 ms, resolution=1x1x1 mm3) with eight different evenly spaced RF phase increments. Imaging was prospectively accelerated using two and 3-fold uniform undersampling in both phase-encoding directions with a fully sampled 50×50 auto-calibration region (ACS). A fully sampled acquisition was also retrospectively undersampled (R=1x8) using 2D CAIPIRINHA and interleaved disjoint undersampling patterns (Figure 1) to ensure unique undersampling per acquisition [6]. Reconstruction was then performed using the CG-SENSE algorithm and g-factor was calculated in all pixels to assess quality [7].
The proposed physics-based method introduces an additional step in the reconstruction pipeline to account for rapid bSSFP signal variations. First, Low-resolution off-resonance maps were estimated from the ACS using the PLANET method [8, 9]. Then, the maps were interpolated to full imaging resolution and incorporated into the bSSFP signal model to generate profiles that more accurately capture the spatial variations (Figure 2). As shown in Figure 3, the physics-based profile estimation improved image quality compared to the conventional method, as reconstructed images showed fewer aliasing artifacts and improved signal uniformity. In addition, g-factor analysis indicated lower noise amplification using the proposed method in all cases. The method preserved steady-state magnetization after acceleration without introducing eddy current distortions or off-resonance artifacts. Prospective undersampling confirmed these results, achieving up to 70% scan time reduction while maintaining high image quality.
Retrospective undersampling further showed that varying sampling patterns across acquisitions improves results, with interleaved sampling providing a nearly artifact-free reconstruction at high acceleration factors (Figure 4). The proposed profile estimation method improves reconstruction quality by introducing an additional computationally efficient step that requires no extra data acquisition. The estimated profiles provide a more precise representation of rapid signal variations, such as those occurring near bSSFP nulls, which are often missed by conventional low-resolution estimates. These errors can become more pronounced at longer TRs or under specific T2/T1 and flip angle combinations. By incorporating off-resonance maps and the bSSFP signal model, our method outperforms conventional approaches while relying solely on ACS data.
In Figure 3, g-factor values below one appear due to regularization, which can partially improve apparent SNR [10]. Also, the decrease in the calculated g_max likely results from problematic pixels in areas of strong aliasing artifacts, which were subsequently reduced through improved profile estimation.
Undersampling patterns also play an important role in optimizing reconstruction quality. As shown in Figure 4, using distinct patterns for each phase-cycled acquisition improved reconstruction quality. These sampling strategies not only alter aliasing appearance and enhance k-space coverage, but also lead to a better-conditioned reconstruction problem for CG-SENSE. The proposed reconstruction framework enables substantial acceleration of phase-cycled bSSFP imaging while preserving image quality. By incorporating off-resonance modeling, the method achieves up to 70% scan time reduction without compromising reconstruction performance. These results highlight the potential for faster, more robust bSSFP imaging in clinical practice.
Maryam KARGARAN (Halle, Germany), Anne SLAWIG, Oliver SPECK, Volkert ROELOFFS
15:40 - 17:10
#47358 - PG456 Multi-endor and multi-site gradient echo-based characterization of 13C coils for clinical studies.
PG456 Multi-endor and multi-site gradient echo-based characterization of 13C coils for clinical studies.
Radiofrequency (RF) coils are an integral part of the MR technology stack, and their characteristics and differences are important to consider when designing a successful study. Hyperpolarized magnetic resonance spectroscopy (MRS), mainly using [1-13C] pyruvate [1, 2], is driving an increased technical development in 13C MRI imaging [3]. Non-hyperpolarized imaging studies with stable 13C compounds, like glycogen investigations, are also being pursued [4].
13C MRS offers unique metabolic insights that complement clinical diagnostics. Due to the short half-life (~ 30 s [5]) and unrecoverable hyperpolarized 13C signal [6, 7], sequences for hyperpolarized imaging needs to be very fast and RF effective to avoid signal spoiling. Evaluating and comparing MR coils for B1+ homogeneity and B1- sensitivity is vital due to the intrinsically low 13C SNR. Finally, multi-center evaluation is important for robust clinical application, highlighting the need for consensus and standardization in order to assure reliable data comparison [8].
We present a novel QC protocol for clinical 13C coils using a basic and cross-platform compatible Gradient Echo (GRE) sequence (Fig. 1). This was tested and compared to a Chemical Shift Imaging (CSI)-based method proposed by Sanchez-Heredia et al. [9]. The GRE protocol was evaluated against the CSI protocol on two 3T Siemens Biograph mMR PET/MRI systems at Rigshospitalet (RH1 and RH2, Copenhagen, Denmark) and on a 3T GE Discovery MR750 system at Aarhus University Hospital (AUH, Aarhus, Denmark). Additionally, the GRE framework was tested on a 3T GE Premier XT system at Cambridge University (CAM, Cambridge, United Kingdom).
All scans used an ethylene glycol phantom (≥99.0% purity, Merk, Darmstadt, Germany) loaded with 17 g/L NaCl [9]. Flip angle (FA) calibration was done before each experiment. The CSI method (9:36 scan time) is based on spectral analysis of a CSI and a separate noise FID scan [9].
We propose a simpler, image-domain-based method. The SNR is calculated using the mean signal (s) of the phantom and the standard deviation of the background area (b):
SNRGREdB = 10*log10(s ̅/σ(b)) (1)
The GRE sequences were acquired with a slice thickness of 100 mm, averages of 1, 4, 8, 16, 32, a 64x64 matrix size, 400x400 mm FOV, 60 Hz/px BW, 25o FA, 50 ms TR, 14 ms TE. Scans were acquired in axial and sagittal planes and averaged for two measurements per coil.
GRE images were normalized by the background mean (5x5 pixels in each corner). An SNR image was created according to eq. 1. The hottest SNR pixels inside the phantom were extracted both for the GRE and CSI methods. Analysis was performed in Python version 3.9 (Python Software Foundation, Delaware, USA) [10].
After the comparative study between RH and AUH, the GRE method was implemented at CAM using 16 averages (83 s scan time).
The characteristics of the coils tested in this study are described in Tab. 1. Fig. 2 illustrates SNR as a function of averages at RH and AUH (R2 = 0.96), and a summary of GRE (16 avg.) and CSI results is presented in Fig. 3. The SNR measurements of the two methods correlate (R2 = 0.72, data not shown).
At RH, the dual-tuned coils (abdomen, mamma, and flex) show lower SNR compared to the single-tuned 13C head coil. The flex coil at RH shows higher SNR than the other dual-tuned coils. The standard error of the mean (SEM) of the scan planes comparing the GRE, Fig. 3A, and CSI, Fig. 3B, shows less deviation between the scan planes when using the CSI method compared to the GRE apparent across all sites. The novel GRE method used for SNR estimation showed a linear correlation with the CSI method (R2 = 0.72). GRE SNR ranged from 12.0 dB to 17.8 dB, and the CSI method showed a wider range and higher level from 52.3 dB to 64.5 dB due to the longer scan time. The GRE method has the advantage of shorter acquisition time and simplified analysis.
The RH head coil (RH2) showed the highest SNR at 17.8 dB using GRE, likely due to its simpler single-tuned design, like the 17.4 dB SNR seen of the breast coil at CAM. Among dual-tuned coils, flex coils generally had the highest SNR (e.g., 16Ch flex and Patch at AUH, and flex at RH2). The 1.5 dB SNR difference of the head coil at RH is likely due to RH1’s older hardware (amplifier and Faraday cage), reflecting a 7-year system age gap.
The GRE sequence showed a maximum SNR difference of 6.2 dB (116.7 %), while the CSI method showed a 12.2 dB (177.3 %). RH2 showed the highest SNR (Head coil #3 Tab. 1), and AUH, NaCl coil #12 Tab. 1, had the lowest for both methods. A similar pattern of SNR between 13C coils has previously been shown [9]. We introduced a new GRE-based 13C coil characterization method, showed excellent correlation to previous methods and successfully tested at three sites with multiple scanners and vendors. This method provides a fair SNR comparison, making it easy, fast, robust, and free of charge [10].
Emil CHRISTENSEN (Copenhagen, Denmark), Andreas CLEMMENSEN, Esben Soevsoe S. HANSEN, Jonathan BIRCHALL, Mary A. MCLEAN, Christoffer LAUSTSEN, Andreas KJAER, Thomas Lund ANDERSEN
15:40 - 17:10
#47663 - PG457 The effect of k-space sampling scheme on sodium quantification accuracy using external reference phantoms.
PG457 The effect of k-space sampling scheme on sodium quantification accuracy using external reference phantoms.
The most common MRI method of sodium (23Na) concentration imaging quantification is to calculate tissue sodium concentration (TSC) maps using references of known sodium concentrations. The phantoms are placed in the field-of-view (FOV) inside the RF coil in proximity of the anatomical region of interest [1,2]. The reference signal concentrations are known and used to create a signal intensity vs. concentration curve (via linear regression) to map voxel signal intensities to their corresponding sodium concentrations. These external references can experience severe B0 field inhomogeneities causing uncorrectable susceptibility artifacts, along with B1 field inhomogeneities that worsen the signal loss after correction which can introduce error into the TSC map [2-7].
Different k-space sampling schemes have been implemented for 23Na-MRI: gradient-echo (GRE) Cartesian, constant-amplitude 3D radial (CA-3DPR) [8], density-adapted 3D radial (DA-3DPR) [9], FLORET [10], rotated spiral imaging [11], and 3D cones [12]. The goal of this work was to evaluate commonly used sodium sampling trajectories in terms of sensitivity to field inhomogeneities and accuracy of quantification in the calculated TSC map.
Sequences were designed and optimized in MATLAB for optimal SNR [13], and implemented on a 3T MR750 (GE HealthCare, Waukesha, WI). The FID for each trajectory was apodised during reconstruction with a matched-filter for increased SNR. Readout times (TRO) in four DA-3DPR sequences (TRO: 5-20ms) were created and compared. Based on simulation, an optimal TRO of 15ms was used for trajectory comparisons (CA-3DPR, Cartesian, FLORET, Spiral, Cones). To investigate steady-state free precession (SSFP) effects, the 15ms DA-3DPR was modified by removing the spoiler gradient. All sequences were designed to have an 80x80x80-matrix size and were acquired over a 240mm FOV. Each sequence was acquired in ~10 minutes, with more efficient sampling schemes having multiple averages.
A 15cm diameter spherical phantom (3% agar with 15mM NaCl), embedded with six 50mL Falcon tubes (3% agar with sodium concentrations of 30-110mM) was fabricated (Fig.1). The phantom was scanned 16 times with each sequence using a 23Na-tuned 16-rung quadrature birdcage T/R RF head coil (24cm diameter) made in-house, with three external references placed alongside the phantom in the coil FOV. The references consisted of 3% agar in 50mL Falcon tubes of sodium concentrations 30, 45, and 70mM.
For each acquisition the mean signal from each of the three references was extracted and linearly regressed to form a signal intensity vs. concentration curve. The mean signal from each of the 7 target concentration regions within the phantom were then mapped to a concentration using the regressed curve (Fig. 2) and then the residual ([23Na]predicted – [23Na]expected, in mM) for each concentration region was calculated (Figs. 2,3). The median residual was calculated for each target concentration region for each sequence (Table 1). As sequences were optimized for brain tissue, accuracy was also determined in the biological range of 20-70mM [1] (Table 1). The residuals were modelled as a linear mixed effects regression model (LMER) using R [14,15], accounting for repeated acquisitions, and was assessed using ANOVA to determine if trajectory contributed significantly to the variance of residuals. When considering the median residual of the target concentration in all regions (Fig. 3), the sequences in order of most to least accurate are: CA-3DPR, 3D cones, FLORET, Cartesian, rotated spiral, DA-3DPR 5ms, DA-3DPR SSFP, DA-3DPR 10ms, DA-3DPR 20ms, and DA-3DPR 15ms. However, due to some of the sequences having residuals crossing 0, it may be more relevant to report the medians of the |residuals|, given in Table 1. Other than the CA-3DPR sequence dropping from most accurate to third accurate, the order remained unchanged. When considering only biologically relevant concentration regions, the order of accuracy remained similar, but the residuals were reduced. Both sequence (p<0.001) and target concentration (p<0.001) contributed significantly to variance for the LMER residual model. SNR, however, did not (p=0.36). K-space sampling schemes contribute significantly to the quantification error in TSC maps when using external reference phantoms. Evaluating signal intensity vs. concentration curves in commonly used sequences, 3D cones and FLORET schemes indicated highest accuracy and DA-3DPR lowest. Accuracy varies based on target concentration which is relevant to anatomical region. Thus, the most and least accurate sampling schemes may change, depending on what range of concentrations are to be measured. For example, 55mM is the overall average concentration in the brain, and the DA-3DPR sequences perform significantly better with that concentration than the CA-3DPR and Cartesian sequences. However, these results are opposite when considering a lower concentration as seen in prostate (30-40mM).
Cameron NOWIKOW (Hamilton, Canada), Rolf F SCHULTE, Michael VAEGGEMOSE, Michael D NOSEWORTHY
15:40 - 17:10
#47814 - PG458 Free breathing compressed sensing dynamic T1 weighted techniques for the liver : golden radial angle vs extra-dimensional, from phantom to patient.
PG458 Free breathing compressed sensing dynamic T1 weighted techniques for the liver : golden radial angle vs extra-dimensional, from phantom to patient.
Accurate liver dynamic MRI/CT is useful in metastatic or hepatocellular carcinoma liver disease. High temporal resolution is needed for accurate arterial phase evaluation. Breath-hold examinations can be difficult for the patient and may result in non-diagnostic quality, which cannot be repeated. When comparing techniques, dynamic techniques cannot be evaluated pairwise due to the need for real-time contrast agent administration. This study aimed to assess the performance of two compressed-sensing based, free breathing acquisition techniques at 1.5T and 3T on phantom in order to be able to optimize on phantom.
Extra-dimensional (XD) and Golden Radial Angle with Sparsity (GRASP) techniques with equivalent TE and TR were evaluated on a 1.5T and 3T MRI (Siemens Healthineers) across a range of flip angles (5-30°) and various fat saturation/suppression methods on phantom. Distortions, artefacts and contrast ratio’s (CR) were evaluated between temperature corrected 346, 489 and 691 ms T1 inserts but also oil for fat saturation. Signal-To-Noise Ratios (SNRs) were assessed using both a pixel-wise series of measurements (SNRmult) and two acquisition subtraction method (SNRNema1).
A direct comparison on volunteer was made for liver percentage signal uniformity, contrast ratio between liver, muscle, saturated and non-saturated subcutaneous fat but also between regular and irregular respiration for both XD and GRASP (2 repetitions). Patient contrast-enhanced CRs at peak contrast and in the late phase were evaluated for the spleen, arterial input function (AIF) and liver parenchyma in 18 patients (89 MRIs) who underwent combinations of XD/GRASP and 1.5T/3T examinations over time. Pairwise comparisons of techniques, MRI systems, patients and organs were performed using Games-Howell tests with Holm-Bonferroni correction. Differences in coefficients of variation (CV=σ/µ, standard deviation/mean) were evaluated using Levene tests. This study was approved under Ethical Committee CEC-2025-011. Phantom distortions were under 1 mm for both GRASP and XD. A trade-off was observed between artefact intensity and extent between GRASP and XD techniques. Phantom CR was reproducible with a CV of 1.6% for XD and 3.2% for GRASP respectively. SPAIR fat saturation reduced fat signal more efficiently for GRASP (90%) than XD (85%), while standard fat saturation was not statistically significant different between GRASP and XD (34-39% reduction). However, SPAIR came with streaking artefacts for GRASP, reduced SNR, reduced T1 489-691 ms contrast for GRASP and XD by respectively 12% and 4% while standard fat saturation did not significantly reduce contrast in that range. Finally, SPAIR required lower temporal resolution. Across flip angles and fat saturation, with equivalent TE and TR, GRASP showed in phantom slightly better contrast compared to XD, between 6% -15% for 489-691 ms while 14-30% for 346-489 ms T1 value (figure 1).
Phantom signal reproducibility showed a CV of 1.5% for XD and 2.7% for GRASP; however, SNR evaluation using the NEMA1 two-image subtraction method resulted in a 54% and 37% CV for XD and GRASP, due to inconsistent noise assessments. Pixel-wise SNRmult showed higher SNR for GRASP, combined with higher temporal resolution.
Figure 1 shows that volunteer-based forced irregular respiration did not show any significant artefacts for either XD or GRASP. Liver percentage signal uniformity was statistically significant better for XD (90%) compared to GRASP (84%) while liver/muscle contrast ratio and fat saturation was better for GRASP.
Dynamic contrast-enhanced intra and inter-patient CRs showed 15% coefficient of variation for the liver and spleen and 30% for the uncorrected AIF. Contrast-enhanced liver peak- and late-phase CR was statistically significant higher for 3T compared to 1.5T, and highest for the XD/3T combination. In clinic, here was a preference for the XD sequence type with reporting of false negative lesions for GRASP. Phantom results showed equivalent distortions and trade-off metal and air artifacts. While SPAIR could improve fat saturation, this came with important contrast and SNR reductions and loss in temporal resolution. Phantom results indicated better contrast and SNR for GRASP. Patient GRASP texture appeared sometimes “grainy” while XD texture was more smooth. Volunteer results showed that irregular respiration, but without movements, was equally corrected by both GRASP and XD. This could possibly indicate that patient movements, during contrast agent administration, were at the origin of artefacts differently corrected by XD, hence the clinical routine preference for XD. Detailed phantom evaluations, based on T1 values, did not align with patient findings: besides a slight uniformity gain in XD, GRASP performed better in phantom. Patient findings showed improved contrast agent enhancement for 3T/XD, next to in-clinic preference and readability for XD. Further research is required to represent
Maxime HUYGHE, Amine ADJOUD (Lille), Eva BRIGE, Sylvain HAVET, Imen EL AOUD, Frederik CROP
15:40 - 17:10
#47306 - PG459 Real-time FIESTA to visualise brain motion in Chiari malformation type 1.
PG459 Real-time FIESTA to visualise brain motion in Chiari malformation type 1.
Chiari malformation type 1 (CM1) is a common condition (the prevalence is approximately 1% in the general population), although it is often an incidental finding and must therefore be related to symptoms. CM1 is defined as a descent of the cerebellar tonsils > 5 mm below the foramen magnum. However, any symptoms are due to altered cerebrospinal fluid (CSF) flow and compression of nerve structures. The Monro-Kellie doctrine postulates that the sum of CSF, blood and brain is constant [1]. In normal individuals the brain motion at the interface of head and spine is very small (approximately 0.14 mm of the brain stem and approximately 0.40 mm of the cerebellar tonsils) [2]. In CM1, due to crowding of the brain stem and herniating cerebellar tonsils in the foramen magnum, the CSF flow is hindered and consequently the brain structures have to move more in relation to the cardiac cycle and respiration. Detecting this increased brain motion is of value in deciding whether or not to perform surgery. Current methods used, such as phase contrast MRI, all depend on gating, either cardiac or using a peripheral pulse oximeter. Failure to achieve good gating is not uncommon with ECG electrodes or pulse oximeters having to be readjusted. We therefore developed a real-time FIESTA sequence (rtFIESTA) that easily visualises brain motion and avoids any problems with gating.
The rtFIESTA sequence is a standard Cartesian balanced steady state free precision sequence (bSSFP), except that it has been optimised to acquire a burst of eleven repetitions of a slice. Bursts are separated by a (necessary) pause to reduce SAR, which results in a disruption of steady state between image sets (note the periodic disruption of image contrast in Figure 1). A single burst is acquired with a single continuous waveform on each gradient amplifier ensuring a k-space velocity (gradient amplitude) that is always greater than zero. This eliminates any deadtime to reduce TR and banding artefacts. The rtFIESTA sequence achieved a temporal resolution of 5 Hz with an in-plane resolution of 1 x 1 mm (24 cm field of view) and slice thickness of 4 mm. A 24 year-old patient planned for occipito-cervical decompression surgery of CM1 was examined with rtFIESTA in a neutral position, as well as in neck extension. The contrast, spatial and temporal resolution was sufficient, with the cerebellar tonsils seen moving in a vertical motion, and the brain stem moving both horizontally and vertically (Fig. 2). During extension the cerebellar tonsils descended slightly more, and the brain stem was slightly more anteriorly positioned, both still moving (Fig. 3). Assessing motion is easily performed as videos in PACS (Picture Archiving and Communication System). Since CM1 is a common incidental finding, information reflecting the dynamic consequences of CM1 is of importance for the clinician. Phase-contrast MRI can be used, but the degree and direction of motion, as well as the degree of compression of nerve structures is difficult to interpret. FIESTA visualizes the brain clearly and by using rtFIESTA any problems with gating is avoided. The scan time is minimal and the rtFIESTA is easily and immediately analysed in PACS as a video. This enables imaging the patient in different positions of the neck (flexion, neutral or extension), during inspiration, expiration, breath-hold, Valsalva manoeuvre, or even during actual movement of the neck. This would add new information for the clinician, since Valsalva manoeuvre and neck extension is known to worsen symptoms [3]. Although motion is only quantified visually, a substantial motion (as in our case) is expected to be of clinical interest, which has been shown with gated FIESTA in symptomatic CM1 patients [4]. rtFIESTA is an easy method to visualise motion of the brain stem and cerebellar tonsils, which is of interest in patients with Chiari malformation type 1. It can also visualise motion during provocation, such as Valsalva manoeuvre and neck extension, which could add new information to whom should be operated on or not.
Skorpil MIKAEL (Stockholm, Sweden), Henric RYDÉN, Adam VAN NIEKERK
15:40 - 17:10
#47672 - PG460 Servo navigation for prospective head motion correction in structural imaging (MPRAGE and 3D-TSE).
PG460 Servo navigation for prospective head motion correction in structural imaging (MPRAGE and 3D-TSE).
Head motion in MRI remains a challenge and can cause artifacts that prevent image diagnosis or bias quantitative measures (e.g. cortical gray matter volume [1]), especially in clinically relevant cohorts that tend to move more. MPRAGE and 3D-TSE are two widely used structural imaging sequences in clinical routine. Many of the published motion correction methods [2] require external hardware, extensive calibration, or can only be conducted retrospectively. Servo navigation has proven to be a marker-free prospective motion correction (PMC) method requiring minimal calibration, short acquisition and offering high tracking precision in steady-state GRE sequences [3,4,5,6]. This work extends the method to the above mentioned non-steady-state sequences.
Five rapid repetitions of an orbital navigator k-space trajectory (400 rad/m, 2.3ms) were inserted before each inversion pulse (MPRAGE) and RF excitation (3D-TSE) (Figure 1). Each repetition included a small excitation pulse (3°) and spoiler gradients; three additional dummy repetitions were played out before the five navigators. This navigator train was used to accelerate motion prediction convergence by updating the scan geometry between navigators (via libXPACE[7] as described in [5]). The linear perturbation model [3,4] was calibrated by the finite-differences (FD) method [3] during the very first navigator train, acquiring three navigators with rotations around x, y, and z, respectively, followed by two unrotated reference navigators. Phantom experiments were conducted to test the stability and step response of the servo control for two calibration methods (FD vs. projection (PROJ) [3]) under 5° angular or 5 mm translational perturbations applied to the navigator orientation.
Two healthy subjects were scanned using a MAGNETOM 7T Plus scanner (Siemens Healthineers, Germany) equipped with a 32 channel Rx (8Tx) head coil (Nova Medical Inc, USA). The first subject was instructed to perform a single large motion during k-space center acquisition of the second volume of a 3D-TSE series (0.5mm iso., TR=3s, TE=504ms, TAvol=5:30 min, GRAPPA 2x2).
A similar experiment was repeated with the same subject for an MPRAGE sequence (0.8mm iso., TR=3s, TI =1.1s, TAvol=2:06min, GRAPPA 2x2). For the second subject, a high-resolution MPRAGE (0.5mm iso., TR=3s, TI=1.1s, TAvol=11:12 min, GRAPPA 1x2) was acquired without instructed motion.
Each scan was repeated without PMC, but with navigators still included to allow for motion estimation. To identify differences in small arteries of the high-res. MPRAGE scan, maximum-intensity projections (MIPs) were calculated (15 cm slab after registration). Fig. 1 shows phantom results, comparing servo control convergence under artificial perturbations of individual motion parameters. Although both calibration methods demonstrate rapid convergence within a few iterations, the FD method exhibits larger within-train oscillations. Notably, FD convergence slows when the field of view (FOV) is rotated (e.g., Rx). Hence, the PROJ method was used in the following experiments.
Fig. 2 demonstrates clear improvements with Servo PMC under an instructed large motion in the 3D-TSE sequence. Moreover, even without instructed motion (baseline), a reduction of ringing artifacts is noticeable in the coronal zoom in the corrected scan.
Fig. 3 presents the results of the MPRAGE motion experiment. As before, Servo PMC yields a clear reduction in artifacts, although it does not fully match the image quality of the reference scan without motion.
Fig. 4 presents high-resolution MPRAGE images. Although differences in the magnitude images are subtle, the MIP of the corrected scan shows improvements in the visibility of small arteries. Oscillations observed in navigator trains of the FD model prediction may result from signal evolution across calibration shots, whereas the preferred PROJ method solely relies on two reference scans.
Residual artifacts in all motion experiments may be exacerbated by fast motion, not corrected due to relatively sparse updates. For MPRAGE, this effect could potentially be mitigated by shifting the navigator train closer to the readout, i.e., after the inversion pulse. However, motion occurring during the imaging train would remain uncorrected. If large pose changes between TRs are detected, reacquisition of corrupted lines at the end of the scan could be considered. Nevertheless, the reduction of motion artifacts in high-resolution images without instructed motion underlines the strong potential of this approach for (ultra-)high-resolution imaging [6].
In the future, robustness against B0 changes could be increased by extending the model to include first-order shim terms [4], which may help to reduce parameter bias. The clear reduction of motion artifacts in instructed motion experiments using servo navigation highlights its strong potential for motion correction, even in non-steady-state sequences such as MPRAGE and 3D-TSE.
Matthias SERGER (Bonn, Germany), Malte RIEDEL, Rüdiger STIRNBERG, Nicolas BOULANT, Klaas PRUESSMANN, Tony STÖCKER, Philipp EHSES
15:40 - 17:10
#47760 - PG461 Feasibility of self-navigated motion resolved 3D lung MRI on a non-commercial 100 mT system.
PG461 Feasibility of self-navigated motion resolved 3D lung MRI on a non-commercial 100 mT system.
Beyond standard spirometry methods for measuring global respiratory function, intensity-based and non-intensity-based proton MRI methods make it possible to map pulmonary dysfunction across the free-breathing lung [1-4] . At conventional magnetic field strengths these approaches require fast acquisitions to overcome inherently short T2* within the lung parenchyma. In contrast, at lower magnetic field strengths, longer T2* values reduce the need for short echo times [5]. The feasibility of an efficient 3D acquisition has been recently demonstrated at 0.55 T, with scan time of 5-minutes facilitated by high performance shielded gradients at 45 mT/m amplitude and 200 T/m/s slew rate [6].
In this study we investigate an alternative imaging regime, examining whether the inverse relationship between T2* and field strength can be used to mitigate the need for high gradient performance for lung imaging performed at low field. Using a non-commercial 0.1 T resistive whole-body scanner, we examine the feasibility of a 3D centre-out radial imaging approach using long readout times to minimise gradient strength and slew rate. We hypothesise that accurate motion resolved lung images can be obtained using gradient performance that is applicable to emerging ultra-low-field MRI technologies below 0.1 T [7-8].
Imaging experiments were performed using a non-commercial, 0.1 T resistive whole-body scanner, equipped with gradients capable of achieving a maximum amplitude of 16.5 mT/m and slew rate of 13.2 T/m/s (Fig. 1). A 3D unbalanced gradient echo sequence was used to acquire 60,000 centre-out radial trajectories, with TE/TR of 0.67/14 ms, voxel size of (3.1 x 3.1 x 3.1) mm3, AZTEK radial trajectory pattern of 1-twist, 1-shuffle, 4-speed [9], and scan time of 14 minutes. Experiments were performed at 4.293 MHz, and 128 readouts were acquired over 10.7 ms. Maximum gradient strength values for pre-phase, readout, and spoiling were 0.62, 0.36, and 5.69 mT/m respectively, with corresponding max slew rates of 3.09, 1.79, and 7.12 T/m/s.
To examine efficacy for motion resolved imaging, data was collected from both a stationary and moving test phantom object, with the scanner bed manually displaced with an amplitude of 30 mm and approximate period of 5 s [9]. In addition, lung imaging was performed on two healthy volunteers instructed to follow consistent slow breathing patterns. Two consecutive acquisitions were performed to obtain a total of 120,000 radial trajectories during a 28-min scan time. A soft-gating approach was implemented and adapted to resolve 16 motion states based on processing of filtered magnitude DC signal variation [10]. For lung and phantom each motion state contained 20,000 and 15,000 radial trajectories respectively. A simple gridding-based reconstruction scheme was deployed using MATLAB, and the utility of the chosen AZTEK trajectory pattern to yield uniform coverage of k-space after soft-gating was quantified by uniform coverage metric [9]. Values ≥ 1 indicate as good or better coverage than randomised radial spoke distribution. The periodic movement of the phantom and lung was observable as a periodic variation of the DC magnitude signal (i.e., the centre of k-space), and 16 motion states were resolved to separate inspiratory and expiratory phases (Fig. 2 – 3). A maximum displacement of (61.0 ± 1.8) mm was measured for the moving phantom (Fig. 4). A total displacement of (25.8 ± 1.8) mm and (17.6 ± 1.8) mm was measured between the inspiration and expiration images for each volunteer (Fig. 4). Use of the AZTEK trajectory pattern yielded uniform coverage of k-space across motion resolved states, with uniform coverage values of 1.01 ± 0.01 and 1.03 ± 0.01 for each volunteer scan. Developing new ultra-low field MRI technologies that target specific applications is one solution towards addressing the challenge of MRI accessibility. Whilst the efficiency of MRI is dependent on accelerating acquisition of k-space, the feasibility of lung imaging at ultra-low field strength is further dependent on addressing the challenges of maximising inherently low values of SNR, field inhomogeneity [11], and minimising errors caused by limited gradient performance. In this study, relatively high values of T2* expected at low field were utilised to allow for longer sampling times and reduce demand on maximum gradient strength and slew rate. Future work will examine the efficacy of alternative imaging approaches to shorten acquisition time and the use of more advanced reconstruction approaches. The feasibility of self-navigated motion resolved 3D lung imaging was demonstrated at 100 mT using gradient performance applicable to emerging ultra-low-field MRI technologies.
Nicholas SENN (Aberdeen, United Kingdom), Gabriel ZIHLMANN, Mathieu SARRACANIE, Najat SALAMEH
15:40 - 17:10
#47895 - PG462 Can PET acquisitions be shortened using an MR powered motion correction framework on a hybrid PET-MRI scanner?
PG462 Can PET acquisitions be shortened using an MR powered motion correction framework on a hybrid PET-MRI scanner?
In clinical practice, liver positron emission tomography (PET) scans often suffer from motion artifacts due to the extended acquisition time and the patient’s free-breathing state. These motion-induced distortions hinder accurate assessment of therapeutic response, necessitate high dose and long acquisition times to achieve sufficient dose deposition and hence SNR.
In this study, we evaluate a shortened PET acquisition (3 minutes 2 seconds) combined with an MR powered 3D non-rigid motion correction framework for PET, leveraging non-rigid motion data derived from a simultaneously acquired, free-breathing T1 Dixon sequence. Our approach builds upon the methodology described in [1], with adaptations tailored specifically for liver imaging.
We acquired simultaneous PET and MR images while monitoring the patient's breathing through a respiratory cushion and the iNav in the T1 motion corrected sequence (Figure 1).
Our processing starts by aligning temporally the raw PET and MRI data streams and truncating the PET data to match the MR acquisition during which the advanced motion monitoring using iNav[2] has been applied. Ultimately, the iNav data enables the calculation of detailed 3D motion fields of liver displacement throughout the respiratory cycle at 4 different states.
Using a single attenuation map (μ-map) generated from a breath-hold sequence (Figure 2.A), we conduct a non-rigid registration between this static μ-map and the MR image (Figure 2.D), which has been resampled to align with the PET image. This step addresses potential misalignments caused by imperfect breath-holds or instances where the breath-hold position does not accurately reflect the expiration phase. The reversed motion fields (Figure 2.F) are now applied to the μ-map to generate μ-maps for each respiratory state (Figure 2.C), enabling precise attenuation correction across different respiratory states.
Using the respiratory motion curve obtained from the iNav (Figure 2.G) and the truncated PET data (Figure 2.H), we now perform respiratory binning of the PET data based on the iNav respiratory signal (Figure 2.I). For each respiratory bin (Figure 2.I), we apply the motion-adjusted attenuation map (μ-map) (Figure 2.C) to enhance attenuation accuracy.
The different attenuation-corrected PET images (Figure 2.J) are subsequently aligned to the expiratory state by applying the corresponding motion fields (Figure 2.G) to each bin. This leads to a summation of the different signals within the final resulting image (Figure 2.K).
Details of the MRI protocol for imaging liver motion has been described previously [3]. In short, the iNav acquired between the T1 sequence blocks data enables the calculation of detailed 3D motion fields allowing us motion solve the image. Figure 3 shows two 3-minute and 2-second PET scans: on the left, a scan obtained without motion correction (Figure 3.A), and on the right, a scan with motion correction (Figure 3.B). When comparing the signal ratio (tumoral liver)/(non-tumoral liver), we observe a 38% difference (6.1 for the motion-corrected PET image versus 4.4 for the non-corrected image). This suggests a clear reduction in noise within the non-tumoral liver tissue. Our concept of using MRI iNAV for correcting PET images shows an improved PET signal, indicating that the motion correction framework effectively recenters the PET signal to its true origin in space. The gain is PET signal within the same acquisition time can – as usual – be played in different directions: firstly, it could be possible to reduce the dose while keep acquisition time, or shorten acquisition time and adjust accordingly dose to recover the same or improved SNR. This improvement in image quality suggests promising potential for more accurate characterization of tumors, intertumoral heterogeneity, potentially enhancing the understanding of therapeutic response and enabling more effective patient stratification.
Currently, we are using only the truncated PET signal aligned with the MRI acquisition (see Figure 1). In the future, we plan to integrate respiratory PMU data from the raw PET signal, extending motion correction across the entire PET acquisition. This enhancement is expected to yield more precise results and improve the overall patient outcome. In conclusion, we have developed an MRI-based PET motion correction pipeline for liver imaging that shows significant potential to enhance the quality of abdominal PET scans. This advancement paves the way for the next phase of our project, where we plan to extend the precise respiratory guidance provided by the iNAV across the entire PET acquisition period, allowing for even higher-quality images.
Our novel PET motion correction framework for liver imaging holds promise for more streamlined patient care and earlier, more accurate treatment assessments
Jake PENNEY (Paris), Khalid AMBARKI, Patrick LEHMANN, Aurélien MONNET, Ricardo SARTORIS, Valerie VILGRAIN, François ROUZET, Kaya DOYEUX, Hatem NECIB, Rene BOTNAR, Claudia PRIETO, Ralph SINKUS
15:40 - 17:10
#47915 - PG463 Reducing motion artefact in high resolution 7T scans using MR MinMo a new head stabilization device.
PG463 Reducing motion artefact in high resolution 7T scans using MR MinMo a new head stabilization device.
High-resolution brain MRI at 7T can significantly improve the detection of pathology[1]. However, motion sensitivity is enhanced due to the increased resolution and scan duration hence even small movements may produce visible artifacts. We therefore aimed to evaluate the effectiveness of the MR MinMo (MR Minimal Motion) head stabilization device[2] in mitigating motion artifacts for high-resolution 7T scans.
To reduce artifacts, retrospective motion correction can also be effective, but it is made more challenging by concomitant interactions with the B0 field[3] and B1 field[4] that are increased at 7T and difficult to correct for larger movements. We therefore additionally tested if there was a synergistic interaction between the use of a retrospective motion correction method called DISORDER[5] and the MR MinMo.
Finally, we examined T2* maps as an application with high sensitivity to bulk head and physiological motion at 7T.
The MR MinMo (Fig1A) is designed to reduce motion in awake subjects aged 6 and older. During the experimental setup, the device was seated into the head coil in the open configuration and participants were position in the MR MinMo.
19 healthy volunteers (7 paediatric aged 10-15yrs, 12 adults aged 20-36yrs) comprising 10 males and 9 females were imaged sequentially with the MR MinMo and standard padding. The order of using MR MinMo versus standard padding was randomized to offset potential order effects[6]. A 2×2 factorial design (Fig1B) was used to evaluate the MR MinMo in four experimental conditions: A1) MR MinMo with linear sampling, A2) MR MinMo with DISORDER sampling and motion correction B1) standard padding with linear sampling and B2) standard padding with DISORDER sampling and motion correction.
Data was acquired using an optimized multi-echo GRE protocol[7] (FA=36°, TR=30ms, TE1-TE10=2.27-26.39ms, BW=470Hz/px) at 0.6mm³ resolution with FOV=256×173×218mm³. Two sampling paradigms were employed:1) a 10-minute scan with linear Cartesian sampling (IPAT=2×2, acceleration factor=4), and 2) a 20-minute scan using DISORDER sampling (acceleration factor=1.4x1.4), where k-space samples are acquired in a pseudo-random order from rectangular regions (Fig1B).
Images were assessed qualitatively by visual inspection of the 1st echo and the T2* map calculated from all echoes and quantitatively using the normalized gradient squared (NGS) metric [8] within a whole-brain ROI. Statistical analysis was performed using a repeated measures ANOVA and T-tests. HV's motion states were estimated within the DISORDER framework with translation and rotation values calculated across orthogonal directions. Maximum values of motion were derived across states then averaged by group(adult vs children). The MR MinMo reduced motion artifacts irrespective of sampling type. Fig2A-B show images demonstrating improved image quality for Mr MinMo using linear sampling. Fig3A-B show the same for DISORDER sampling with and without motion correction. Motion state analysis showed paediatric subjects exhibited substantially higher motion without the MR-MinMo compared to adults. For children vs adults, group average of the max. translation was 0.26mm vs 0.09mm and for rotation 0.15° vs 0.04°. The NGS values are shown in Fig.4, a significant main effect of using the MR MinMo (p=0.002) was found. A paired t-test confirmed that retrospective correction was effective in improving the quality of the DISORDER sampled scans. However, these were not significantly better than the linear sampled equivalents (p=0.48). A significant interaction was found between the MR MinMo and DISORDER motion corrected scans (p=0.02). An interaction between MR MinMo and age (p=0.02) suggested greater efficacy in children who generally exhibited more motion.
T2* maps calculated from linear sampled echoes showed improved uniformity and reduced motion corruption with the MR MinMo (Fig2C). The MR MinMo demonstrated a significant improvement in image quality both with and without retrospective motion correction demonstrating its potential to work synergistically with other motion correction approaches. We speculate that this is because the reduced motion range may lead to greater data consistency with algorithmic assumptions. As expected, younger subjects tended to move more as measured using the estimated motion states. The randomization of scan order across subjects ensured that any increase in motion prevalence with total exam duration [6] did not affect results. This approach also doesn't require additional hardware or custom manufacturing, making it suitable for routine use. The MR MinMo is an effective solution for reducing motion and increasing image quality in high resolution 7T scans. The device is compatible with and may improve the performance of retrospective motion correction methods.
Jyoti MANGAL (London, United Kingdom), Simon RICHARDSON, Yannick BRACKENIER, Matthew GARDNER, Pierluigi DI CIO, Chiara CASELLA, Shaihan MALIK, Jo HAJNAL, Martina CALLAGHAN, Fred DICK, David CARMICHAEL
15:40 - 17:10
#47647 - PG464 Monitoring head exposure around a 14.1 T preclinical MRI scanner using smart goggles equipped with magnetometers.
PG464 Monitoring head exposure around a 14.1 T preclinical MRI scanner using smart goggles equipped with magnetometers.
Throughout their workday, the staff operating MRI equipment are continually subjected to its electromagnetic fields, which include static field, radiofrequency (RF) emissions, and gradient fields. Exposure to static magnetic fields (SMF/B0) is known to cause several effects, such as a metallic taste in the mouth, nausea, nystagmus, and vertigo [1]. Gradient fields have the potential to stimulate peripheral nerves, whereas RF signals are primarily associated with heating of body tissues [2]. This research work emphasizes only the exposure to SMF, which is commonly assessed using portable exposimeter currently available [3]. These exposimeters are typically attached to the chest, leading to a notable difference between the magnetic field measurements, and the magnetic field levels experienced at the head level. Moreover, the variance in movement between the chest and the head adds complexity to the analysis of gradients related to motion around the magnet. In response to this complexity, we have developed smart goggles equipped with multiple magnetic field sensors over the past few years [4-5]. These smart goggles offer greater accuracy compared to pocket exposimeters and are designed to be performant and comfortable enough to make them suitable for studies involving cohorts of MRI workers working with different scanners.
The smart goggles used in this work have been presented in [5]. It features ten sensors at locations near the ears, the temples, and the eyes. The goggles feature the SENM3Dx sensors (Senis AG), capable of measuring ± 4 T on each axis (Fig.1). The exposimeter samples magnetic field signals at 47 Hz and is connected to a non-ferromagnetic battery powered data logger that stores data on an SD card. In the subsequent measurements, the magnetic field norm B is determined using the Euclidean norm. The slew rate is computed individually for each of the sensors by taking the time derivative (i.e., dB/dt) of B. The gradient is calculated as the difference between the norms of two opposing sensors located near each temple, assuming a distance of 17.5 cm between the sensors (Fig.1). For this work, the volunteer researcher moved around a 14.1 T preclinical scanner (Varian, Burker). He was tasked to carry out routine activities in the MRI room. These activities involved simulating the setup of a head antenna, which entails bending down and looking into the bore with the head roughly aligned with the center of the bore at its entrance (Fig.2), as well as moving around the scanning bed. The findings from the measurements are illustrated in Fig.3. The data reveals that the measured magnetic field norms reached a maximal value of 2.3 T. The observed slew rates peaks at -3.7 T/s. Additionally, the maximum calculated gradient field was determined to be 2.3 T/m. These results indicate that during these measurements, exposure levels surpassed the thresholds of 2 T, and 2.7 T/s established by the laboratory-related safety hazards directive 2013/35/EU [6]. Preclinical MRI workers are exposed daily to high static magnetic fields, yet the origin of the physiological effects of such exposure remain largely unstudied. In this study, we have highlighted the significant magnetic field dynamics encountered by these workers as they move around a preclinical scanner operating at 14.1 T. By involving a substantial number of participants who interact with various MRI models and protocols, we aim to gain deeper insights into the physiological impacts of exposure to B0 generated by MRI scanners. Conducting broad investigations will be crucial for understanding the mechanisms that lead some individuals to experience SMF-related effects. In the long term, these studies could pave the way for defining new safety recommendations specifically tailored for individuals subject to these effects, ensuring better protection for MRI workers. In this study, we used the smart goggles developed in [5] to focus on SMF exposure of preclinical MRI workers. In the preclinical context, the magnetic fields are often stronger than those encountered in clinical settings, although the MRI bores are significantly smaller. Our objective was to assess whether their exposure levels were comparable to those we had previously measured [5]. This is the case and the measured exposure levels can exceed recommended thresholds. The results obtained are similar to those observed for a 7 T whole-body MRI scanner, which does not have any counter-field coils. Further investigations will be necessary to understand the mechanisms that cause some individuals to experience SMF-related effects. Prior to these further investigations, we plan to enhance the ergonomy and user-friendliness of the exposimeter.
Thomas QUIRIN, Hugo NICOLAS, Corentin FÉRY, Nicolas WEBER, Julien OSTER, Jacques FELBLINGER, Joris PASCAL (Muttenz, Switzerland)
|
Poster hall |
17:10 |
TIME FOR A BREAK
- Coffee and refreshments will be available at the cash bar.
|
17:30 |
"Friday 10 October"
Added to your list of favorites
Deleted from your list of favorites
A26
17:30 - 18:30
HOT TOPIC ROUND TABLE
Are we redundant? The future of radiology professions
17:30 - 18:30
Survival strategies for MR physicists.
Moritz ZAISS (Keynote Speaker, Germany)
17:30 - 18:30
Survival strategies for radiographers.
Jonathan MCNULTY (Keynote Speaker, Ireland)
17:30 - 18:30
Survival strategies for radiologists.
Marion SMITS (Keynote Speaker, Rotterdam, The Netherlands)
|
Auditorium 900 |
18:30 |
POSTER DRINKS
|
Saturday 11 October |
08:30 |
"Saturday 11 October"
Added to your list of favorites
Deleted from your list of favorites
A30
08:30 - 09:30
FT1 Oral - Low Field Technology
From physics to the clinic
08:30 - 08:40
#47804 - PG031 In vivo imaging with a low-cost MRI scanner in low-resource settings.
PG031 In vivo imaging with a low-cost MRI scanner in low-resource settings.
Lack of access to essential medical imaging is a prominent burden on Low- and Middle-Income Countries (LMICs). Low-field MRI (LFMRI) technologies have been postulated to alleviate this [1]. However, a major hurdle with these systems is electromagnetic interference (EMI), i.e. unwanted noise caused by external sources and picked up at the receive (Rx) chain. As the number of available LFMRI systems increases, we often encounter that these scanners underperform due to low signal-to-noise ratio (SNR) caused by EMI.
Challenges are amplified in LMICs, where electric power lines tend to be unreliable and components and equipment scarce. Indeed, reliable imaging with MRI scanners constructed on site in LMICs remains a pending task.
In this work, we demonstrate significant improvements to an existing scanner built at Mbarara University of Science and Technology (MUST), Uganda, which enabled us to acquire the first in vivo images using a low-cost LF-MRI system in Africa.
The MUST 50 mT system is the first MRI device constructed in Africa [2]. We have revamped the system to enable in vivo image acquisition. Specifically, we have acted on the electronics, cabling, and scanner console.
Electronics and cabling. We minimized EMI by shortening and separating cables by function (RF, gradients, power, digital), with shielding—especially for gradient lines near the antenna. RF components and EMI sources were boxed and grounded, following a star-ground layout referenced to the RF shield. Despite unavoidable ground-loops through the mains, all subsystems were powered from a single outlet connected to an uninterruptible power supply with a 50 Hz filter. Scanner noise was assessed via RMS voltage measures at the Rx coil, expected near 174 dBm/Hz, i.e. the thermal (Johnson) noise floor of a 50 Ohm resistor at room temperature.
Control system. We updated MaRCoS [4] and MaRGE [5] to their latest stable versions and compared system performance against a commercial spectrometer (Magritek KEA2). The latter is significantly more expensive but can serve as a noise floor benchmark. We operated in four different configurations: MaRCoS with the MUST LNA and TxRx switch (MaRCoS + MUST); MaRCoS with the KEA2 duplexer electronics (MaRCoS + DUP); KEA2 + MUST; and KEA2 + DUP.
The implemented upgrades made in vivo acquisitions possible. We obtained T1-weighted RARE images of the knee and brain (sequence parameters in Figs. 3-4). The revamped system is shown in Fig. 1. To quantify the improvements, Fig. 2a presents noise measurements under various conditions: optimized for EMI, unoptimized, using a 50 Ohm resistor, and when controlled with the KEA2. Figure 2b illustrates the impact of different control systems and receive electronics on image quality.
Finally, we present the in vivo images acquired: Fig. 3 shows a knee scan and Fig. 4 a brain image, both from healthy volunteers. A key focus of this work was the characterization and mitigation of system noise. With the Rx coil and matching electronics, we achieved 2.3× Johnson with MaRCoS, which aggravates if we do not handle EMI adequately and depends on external usage of the building power grid. The scanner is particularly sensitive to the MaRCoS housing, which is not fully connectorized due to the lack of basic components. Note that this is not a fundamental limitation [3] and shall be improved shortly.
The choice of control system also had a substantial impact on system performance, usability, and cost. MaRGE enabled noise measurements, calibrations, and image acquisitions, greatly enhancing user experience. With KEA2, we were able to operate at 1.7× Johnson, suggesting we have room for improvement with MaRCoS. In any case, despite the slightly higher noise observed with MaRCoS in this setup, the resulting images exhibit comparable quality, underscoring its practical viability.
Lastly, the image quality is consistently sufficient for in vivo acquisitions. In the brain image, we observe a dark area at the bottom, which is due to the excitation bandwidth not being sufficient to cover the entire frequency spectrum due to magnet inhomogeneities. This could be remedied by using higher RF power or working on a shimming system to improve the homogeneity of the main magnet. This last point is key to continuing to improve the system. With a more homogeneous magnet, we will be able to decrease the acquisition bandwidth without observing major distortions, reducing noise, and therefore increasing the SNR allowing us to reduce the image time. In conclusion, we present the first in vivo images acquired using a low-cost MRI system in Africa. This required substantial improvements to the MUST system and overcoming the additional challenges posed by operating in a low-resource setting. Our work represents a first step toward the clinical translation of low-field MRI systems in LMICs, with the potential to significantly improve access to medical imaging.
Teresa GUALLART-NAVAL (Valencia, Spain), Ronald AMODOI, Mary A. NASSAJJE, Robert ASIIMWE, Patricia TUSIIME, Maureen NAYEBARE, Leo KINYERA, Lemi ROBIN, Patience NINSIIMA, Faith NATUKUNDA, Joachim MUSIIMENTA, Heaven NAMBI, Florence NAMAYANJA, Benjamin WAMONO, José Miguel ALGARÍN, Thomas O'REILLY, Andrew WEBB, Steven J. SCHIFF, Johnes OBUNGOLOCH, Joseba ALONSO
08:40 - 08:50
#47618 - PG032 3D ULF MRI at 10 mT Enabled by Ultrasensitive SQUID Detection and High Homogeneity in an Open Environment.
PG032 3D ULF MRI at 10 mT Enabled by Ultrasensitive SQUID Detection and High Homogeneity in an Open Environment.
Recent advancements in ultra-low field (ULF) MRI systems operating at or below 10 mT present a transformative approach to reducing system costs and expanding accessibility while mitigating traditional MRI contraindications such as claustrophobia and metallic implants [1-4]. Enhanced T1 contrast in low-field regimes provides unique diagnostic opportunities [5], yet low signal intensity proportional to B0 has historically limited clinical applicability due to diminished signal-to-noise ratio (SNR). To address these limitations and make MRI more affordable and accessible, we developed a 10 mT MRI system that combines an ultrasensitive detection system using superconducting quantum interference devices (SQUIDs) coupled with a room-temperature volume gradiometer for enhanced sensitivity and with advanced denoising and reconstruction techniques [6,7]. Additionally, a resistive Merritt coil electromagnet provides B0 homogeneity superior to conventional permanent magnet systems that typically exceed 200 ppm and can reach 10,000 ppm in Halbach configurations [4,8,9]. This combination allows full-body imaging capabilities including a vast possibility in the choice of sequences that are not feasible with other low-field MRI systems. In this work, we present 3D contrast imaging of a phantom with varying concentrations and a garlic bulb at 2.5 × 2.5 × 10 mm³ resolution, demonstrating the performance of our SQUID-based MRI system for high-quality imaging in an open environment.
Our MRI system, depicted in Figure 1, operates at 10 mT using a Merritt coil electromagnet for B0 generation with stability maintained at <100 ppm through active field compensation. The RF transmit saddle coil, consisting of five turns, is tuned to 426 kHz. Signal reception is performed by a 20-turn room-temperature volume gradiometer coil connected to a low-Tc niobium SQUID operating in current sensing mode (Figure 2). The SQUID, housed in a cryogen-free cryostat and cooled to 4.2 K using a pulse tube cryocooler, achieves a 2 nA/√Hz noise floor. The system is enclosed in an aluminum shield to mitigate electromagnetic interference (EMI). Gradient coils generate a 125 µT/(A·m) gradient strength across three orthogonal axes, and three-axis shimming maintains B0 uniformity. Noise suppression is further enhanced by implementing the EDITER method [10]. 3D images of a contrast phantom with varying MnCl₂ concentrations and a garlic bulb were acquired using spin echo (SE), gradient echo (GRE), and inversion recovery GRE (IR-GRE) sequences (Figure 2). Optimized imaging parameters derived from T1 and T2 measurements facilitated the acquisition of 3D contrast images (Figure 3). The GRE sequence provided T1-weighted and proton density-weighted images, the SE sequence produced T1-weighted and short tau inversion recovery (STIR)-like images, and IR-GRE sequences generated STIR and fluid-attenuated inversion recovery (FLAIR) images. The acquisition times varied, ranging from 15 minutes to 1 hour, depending on the sequence parameters. In addition to phantom imaging, we acquired 3D images of a garlic bulb at a spatial resolution of 2.5 × 2.5 × 10 mm³ using the T1-weighted GRE sequence, demonstrating the system’s capability for resolving structural details in biological samples at 10 mT (Figure 4). This work presents a novel 10 mT MRI system integrating an ultrasensitive SQUID detection system and a highly homogeneous B0 field, achieved through a resistive magnet, enabling full-body imaging capabilities. Our system not only reduces operational costs by eliminating the need for cryogenic cooling but also expands diagnostic imaging capabilities. The ultra-low field regime presents opportunities for novel contrast mechanisms and imaging sequences due to enhanced T1 contrast without gadolinium-based agents, which could potentially provide additional quantitative information and diagnostic insights achieved through balanced steady-state free precession (bSSFP) or synthetic MRI techniques [11,12]. Our ongoing work focuses on further enhancing SNR toward clinical applications through optimization of the SQUID-gradiometer interface, developing a cooled receiver coil to reduce Johnson noise, and integrating CNN-based denoising techniques to mitigate environmental EMI. Additionally, k-space trajectory optimization is under investigation to accelerate acquisition times without compromising image quality. We present a cryogen-free, SQUID-based MRI system operating at a highly homogeneous B0 field of 10 mT. This system can acquire 3D images of biological samples with a spatial resolution of 2.5 × 2.5 × 10 mm³. These results provide a promising foundation for future developments in ultra-low-field MRI as an accessible and cost-effective diagnostic modality.
Marco FIORITO, Alexandre JAOUI, Dimitri LABAT, Isabelle SANIOUR (Paris), Mustafa UTKUR
08:50 - 09:00
#47586 - PG033 Nonlinear model-based b0 field compensation in low-field mri using hall sensor arrays and spatial interpolation.
PG033 Nonlinear model-based b0 field compensation in low-field mri using hall sensor arrays and spatial interpolation.
Low-field magnetic resonance imaging (MRI) systems based on permanent-magnets below 0.1 T
are increasingly utilized in portable and resource-constrained settings due to their compactness,
reduced power requirements, and enhanced safety. The performance of such systems is suscepti-
ble to temporal fluctuations in the B0 magnetic field, which can emerge from thermal variations
and environmental disturbances. These instabilities degrade the spatial homogeneity, impairing
image quality. Conventional compensation strategies, including static frequency configuration
and recalibration, are insufficient under dynamic operating conditions. This study introduces
a data-driven approach for continuous B0 drift correction that combines directional dispersion
maps obtained from Hall effect sensors [1,2] with voxel-wise field interpolation, and nonlinear
spatia model predictive control (MPC) [3,4]. The proposed methodology aims to achieve robust,
real-time B0 stabilization without reliance on manual recalibration.
A 3D B0 field mapping was performed within a spherical field of view (FOV) of a 50 mm
Magnet [5]. Continuous, measurements were taken every 10 min over several hours, shown in
Fig. 1. Each field map consisted of 33 points in the FOV. An array of six MLX90393 Hall
sensors was mounted on the outside, along the isocenter of the magnet in a circular arrangement
at an angle of 30◦ shown in Fig. 2. This configuration allows the measurement of the magnetic
field polarization and characterization of the dispersion field, which is on average about 1.5 mT
compared to the 46 mT main field. This stray field emanating from the geometry of the Halbach
magnet is shown in Fig. 3. A nonlinear quadratic regression model was fitted to the 24-hour
sliding window data from the dispersion data of B0 field map. Model predictive and Spatial
multipoint interpolation was used for estimation per voxel B0 value in the FOV. The model achieved a maximum square error (MSE) of 100 μT2, an MAE of 9 μT, and a root
mean squared error (RMSE) of 11 μT on the test set, with a coefficient of determination (R2) of
0.934. These results indicate strong agreement between the model predictions and the measured
B0 field values shown in Fig.4. The model achieved > 99.8% of the variance in the FOV B0 maps
with residuals below 50 μT. Using realtime measurement data, kept the central B0 prediction
errors below 0.1 mT (0.2%) over 24-hour. Combined spatial and temporal estimation yielded voxel-wise B0 predictions with errors <80 μT over the FOV.
The Larmor frequency prediction matched the ground truth and enables continuous RF retuning within 2 kHz (≈47 μT) [6]. For
validation, using the B0 mapping field probe inserted in the isocenter shows a field stabilization
of 0.12 mT, which demonstrates the functionality of the MPC model in this limited validation
setup. The integration of nonlinear spatial modeling, MPC, and directional dispersion field analysis
provides a robust framework for real-time B0 stabilization in low-field MRI. The triaxial Hall
sensor array arranged at 30◦ intervals offers directional sensitivity that allows for both tempo-
ral drift estimation and spatial dispersion characterization. The model’s low prediction error,
supported by sub-10 μT MAE and a strong R2 score, demonstrates that dispersion fields from
Halbach magnet geometries can reliably inform both voxel-wise estimation and active shimming
strategies [1]. While these results are based on synthetic and reference sample validation, further
testing with dynamic imaging sequences is required to assess generalizability under operational
conditions. Remaining limitations include inhomogeneities beyond the modeled volume and the
need for continuous calibration under long-term environmental variation. This study demonstrates that outside directional dispersion fields, in combination with nonlinear
quadratic model predictive control, enables precise (<0.2%) B0 field drift correction in low-field
MRI over 24 hours. The approach supports continuous frequency adaptation and voxel-wise field
estimation with sub-10 μT accuracy. The addition of a closed-loop active drift compensation
driven by the MPC actively enhances stability. These results highlight the potential for robust,
compact MRI systems with minimal calibration requirements.
Marcel Werner Heinrich OCHSENDORF (Aachen, Germany, Germany), Kostiantyn LAVRONENKO, Volkmar SCHULZ
09:00 - 09:10
#46161 - PG034 Hard tissue imaging with ZTE sequences in a portable Halbach system.
PG034 Hard tissue imaging with ZTE sequences in a portable Halbach system.
The MRI community's heightened interest in low-field scanners (B0 < 100 mT) has led to the development of low-cost, lightweight and portable systems. Constrained to these attributes, Halbach arrangements play a role for both musculoskeletal applications [1,6]. In extremity imaging, details from hard tissues like bones, tendons or ligaments (with T2*<1 ms) are of medical interest. Conventional echo sequences lack this capability, but dedicated sequences for short T2 encoding, such as PETRA and other ZTE variations [3], are used clinically at clinical field strengths. However, they are not trivially applicable in low-cost Halbach systems, mostly due to challenging requirements such as high RF power and excellent field homogeneity. In this work, we demonstrate knee imaging with PETRA in our 72 mT Halbach system and report the first in vivo measurements of the T1 of hard tissues at low field.
Experiments were performed in our portable MRI scanner with a 72 mT magnet based on Halbach and conceived for extremity imaging (Fig 1.a) [1]. The B0 field can be shimmed down to 1200 ppm over a 10 cm DSV. The gradients can run stably at 25 mT/m during hundreds of ms. The RF coil employed is a solenoid with 15 cm diameter and 15 cm length. The Radio-Frequency Power Amplifier (RFPA) is rated for up to 500 W. The system is controlled by the open-source MaRCoS console [2, 4], which we have upgraded to include a PETRA sequence (Fig 1.b) [3]. Image reconstructions are performed with ART and employ prior knowledge of the B0 distribution to suppress image distortions by means of our recent Single-Point Double-Shot technique (SPDS) [5].
All images were with the shortest hard RF pulses allowed by the system (<20 us) to ensure a coherent, full bandwidth excitation. Acquisition times were taken to be compatible with the shortest expected T2 times (~1 ms), since this never produced significant gradient heating in the system. The shortest TE achievable is constrained in our setup to TE=300 us, due to long-lived transients generated by switching from transmit (Tx) to receive (Rx) mode.
To estimate T1 values of hard tissues, we acquired a set of PETRA images with constant, short TR < T1 and variable flip angle (FA) to induce an incoherent steady state in all tissues. In this regime, the dependence of SNR on Mo, theta, T1, and TR is exploited to estimate Mo and T1 of every tissue according to SNR=Mo·(1-E1)·sin(FA)/(1-E1·cos(FA)), being E1=exp(-TR/T1). Figure 2a exhibits the performance of PETRA in our Halbach system after a 24-minute scan for a T1-weighted image. Cortical bones, ligaments, and tendons are all visualized brighter than the background noise. However, ZTE sequences are heavily proton-density-weighted and do not offer much anatomical contrast compared to echo-based sequences. Figure 2b compares hard-tissue visualization with PETRA against a conventional RARE sequence with the shortest possible TE in our setup (10 ms).
In Fig. 3a we show a set of PETRA images acquired with TR=50 ms and variable theta. Incidentally, this patient diagnosed with a possible Baker cist, even if this is irrelevant to our study. In Fig. 3b, we show the SNR evolution of each tissue depending on theta, together with fits according to Eq. (1) to determine T1 values. These are: T1,muscle=181.5 ms, T1,lipid=124.4 ms, T1,cortical bone=106.4 ms, T1,spongy bone=99.4 ms, T1,tendon=32.1 ms and T1,ligament=98.9 ms. The fit yields also estimates for the proton densities relative to lipid: Mo,muscle=0.94, Mo,lipid=1, Mo,cortical bone=0.47, Mo,spongy bone=0.83, Mo,muscle=0.94, Mo,tendon=0.91 and Mo,ligament=0.53. We have obtained the first musculoskeletal images showing hard tissues of a knee in a portable, low-cost scanner. This demonstration opens a qualitative new path for low-field MRI. Our quantitative estimations for spin-lattice relaxation times for muscle and lipid at 72 mT are in acceptable agreement with those reported by Webb et al [6] at 50 mT, where they found T1,lipid=130±5 ms and T1,muscle=171±11 ms. Note, however, that we have not found T1 estimations for hard tissues in the knee in the existing literature. Our study shows PETRA imaging is possible even in low-field, portable MRI scanners. This could be relevant for musculoskeletal applications.
The main limitation encountered so far is that scan times with PETRA are rather long, meaning translation to clinical practice may still require significant hardware improvements. In our setup, the main penalty is created by the long delay needed for TxRx switching, enforcing substantial pointwise sampling at the center of k-space.
Borreguero Morata JOSE, De Castro Santhos LUIZ GUILHERME, Fernández García MARINA, Castanón García-Roves ELISA, Vega Cid LORENA, Guallart Naval TERESA, Algarín Guisado JOSE MIGUEL, Galve Conde FERNANDO, Alonso Otamendi JOSEBA (Valencia, Spain)
09:10 - 09:20
#47797 - PG035 An open-source framework for remote data processing from low-field scanners.
PG035 An open-source framework for remote data processing from low-field scanners.
As low-field MRI (LFMRI) scanners and related technologies continue to expand, the computational power provided by local control PCs becomes insufficient for advanced data processing tasks. Some examples include model-based image reconstruction, distortion correction, or tissue segmentation, more so if deep learning (DL) methods are employed, and especially in the context of affordable systems for use in low- and middle-income countries [1,2]. To our best knowledge, existing solutions for remote and cloud data processing are either proprietary or not open to the public (e.g. Cloud-MRI [3]).
Unlike with high-field scanners, which are designed to be vastly multi-purpose, the hardware and software architectures of LFMRI systems are greatly dependent on the targeted applications. This makes adaptable open-source control systems and software solutions an appealing option for the laboratories and spin-off companies manufacturing these devices. Among the available alternatives, MaRCoS stands out for its high performance and versatility [4,5]. MaRGE [6] is the newest open-source graphical user interface (GUI) for MaRCoS, it is programmed in Python, and it has been designed to cover needs from both researchers and clinicians. On the other hand, Tyger is a recently released open-source platform for remote signal processing [7]. With Tyger, the MR data generated by an LFMRI scanner anywhere in the world can be streamed to the cloud (e.g. Microsoft’s Azure cloud), even through mobile networks. Signal processing code can be written in any language, as long as it can read and write to named pipes (which are file-like but do not support random access). There is no SDK, meaning one can develop, test, and debug code locally using only files, without Tyger dependencies. Once finished, a Docker container image can be generated to run the exact same code in the cloud with Tyger, and the results can be piped back to the scanner.
Here we present an integration of Tyger into the MaRCoS and MaRGE ecosystem and we thereby release the first open-source framework for cloud data processing with a focus on LFMRI (Figure 1). With this development, affordable MRI systems sited anywhere in the world can now leverage the most powerful compute available in the cloud for data processing, image reconstruction and, eventually, the generation of diagnostic information.
We have expanded the post-processing window in MaRGE with new buttons for executing specific methods in the cloud through Tyger. At a lower level, the MaRCoS native data files are translated into the open MRD standard [8,9], which is then piped to the cloud, where we run specific reconstructions programmed using simple Python syntax (Figure 2). All code is publicly available on GitHub [10].
We performed two sets of reconstructions of data acquired in two different portable scanners. On the one hand, we used our 84 mT elliptical halbach scanner [11] designed for neuroimaging to test image reconstruction with the aim of correcting distortions due to magnetic field inhomogeneities (B0). These reconstructions include: iterative Algebraic Reconstruction Techniques (ART) for solving the linear set of equations determined by an encoding matrix that can optionally incorporate prior knowledge (PK) of the B0 spatial distribution [12]; and conjugate phase (CP) [13]. In both cases we used the SPDS method [12] to obtain the B0 map. In addition, we include a standard inverse Fast Fourier Transform (iFFT) protocol to compare the improvement.
On the other hand, we work with our portable 72 mT LFMRI system [2] designed for extremity imaging, with which we test the application of deep learning methods computed in Tyger with the aim of improving image quality. We input an iFFT reconstruction of the knee to a Pix2Pix network [14] pre-trained with 160 pairs of knee images acquired in our system and in a Philips Achieva 3T scanner at La Fe Hospital (Valencia, Spain). Figure 3 shows Python reconstructions of a brain acquisition from Tyger with iFFT, ART+PK and CP.
Figure 4 presents an LFMRI knee reconstruction, a 3T image of the same knee, and the output of the Pix2Pix network generated by Tyger. We have demonstrated a successful integration of Tyger with MaRGE, connecting affordable low-field MRI scanners to some of the most powerful computational cloud resources. Importantly, this work is conceived as an illustration of the potential brought along by this integration, so we have merely programmed a few selected applications that include conventional (Fourier) and advanced (compressed sensing, iterative and DL-based) image reconstruction methods. These leverage access to some of the most powerful MRI data processing toolboxes available, including BART [15]. However, this list is far from exhaustive and can eventually include strictly any tool that can be loaded into a Docker image.
Teresa GUALLART-NAVAL (Valencia, Spain), José Miguel ALGARÍN, José BORREGUERO, Fernando GALVE, John STAIRS, Michael HANSEN, Joseba ALONSO
09:20 - 09:30
#47415 - PG036 First Clinical Evaluation of a portable 47 mT Halbach MRI system for Detecting Arthritis-Related Inflammation.
PG036 First Clinical Evaluation of a portable 47 mT Halbach MRI system for Detecting Arthritis-Related Inflammation.
Rheumatoid arthritis (RA) is a chronic inflammatory disease affecting approximately 1% of the global population. Early detection and treatment are critical for improving long-term outcomes and reducing healthcare costs[1, 2]. While expert-led screening clinics could support early diagnosis[3], they are not widely available and often impractical in primary care. MRI is a reliable imaging tool for detecting joint inflammation, offering high sensitivity and specificity compared to ultrasound[4]. However, conventional MRI is costly, often inaccessible and uncomfortable, requiring full-body entry into the bore and tight hand immobilization. Portable low-field MRI has emerged as a promising alternative due to its affordability, comfort, and suitability for decentralized care[5, 6]. In this project, we evaluated a contrast-free, fluid-sensitive MRI protocol using a portable low-field scanner to assess hand inflammation in patients with clinical arthritis, aiming to support earlier, more accessible diagnosis.
All data were acquired using a 47 mT (1.98 MHz) Halbach-based MRI scanner with a Magritek Kea2 spectrometer (Aachen, Germany)[7] and a separate transmit/receive coil (Figure 1).
Sixteen patients with suspected RA were assessed at the Early Arthritis Recognition Clinic (EARC) in Leiden. All patients underwent a targeted physical examination by rheumatologists to determine the presence/absence of clinically apparent inflammatory arthritis in the wrist, metacarpophalangeal (MCP), and proximal interphalangeal (PIP) joints (Figure 1d). In the presence of confirmed clinical arthritis, patients were subsequently scanned on the low-field MRI system while comfortably seated next to the scanner (Figure 1a). We applied a previously developed fluid-sensitive MRI protocol optimized for inflammation detection without contrast agents[10]. It included a T1-weighted turbo spin-echo (TSE) scan for anatomical reference and a heavily T2-weighted, fat-suppressed STIR scan for fluid sensitivity. Parameters were as follows:
T1w: TR/TE = 400/16 ms – scan time approximately 4 minutes (depending on hand size).
IR-T2w: TR/TE/TEeff = 1800/15/75 ms – scan time approximately 12 minutes.
Both scans used a resolution of 1×1×3 mm³ and a bandwidth of 20 kHz. Additionally, the IR-T2w scan was denoised using BM4D[11].
Two experienced clinicians independently assessed the images for joint inflammation using a custom viewer developed in MeVisLab (MeVis Medical Solutions AG, Bremen, Germany).
The diagnostic performance of low-field MRI was assessed by comparing image-based findings to physical examination, considered the reference standard. Sensitivity and specificity were calculated per anatomical region. A flexible scoring criterion was applied: inflammation was recorded if detected by at least one reader.
A subset of patients (n = 10/16) also received a 3T MRI examination (Philips, Best, the Netherlands), using a previously published protocol[9] – these data were used for visual comparison but not for analysis. The custom-built viewer (Figure 2) was essential for image interpretation, enabling intuitive slice navigation and real-time contrast adjustment. Figure 3 shows the image quality achieved across cases. Despite variation in noise levels, inflammation was consistently detectable, even in the most challenging cases. Comparisons with 3T highlight the diagnostic value of low-field MRI in identifying joint inflammation.
Table 1 shows that the diagnostic performance varied across joint levels. Sensitivity was highest in the wrist and lowest in the PIPs, while specificity remained consistently high across all joints (range 88-100%). The system was especially effective in ruling out inflammation, as reflected by high NPVs throughout (range 93-100%). These trends suggest that low-field MRI may be well suited as a screening tool, particularly for reducing unnecessary referrals in cases without joint inflammation. This study represents the first evaluation of low-field MRI using a Halbach-based system for detecting joint inflammation. The results are promising, particularly in terms of high specificity and NPV, supporting its potential as a screening tool. However, during the study, we identified few areas for improvement. The relatively low sensitivity observed in the PIPs reflects suboptimal initial positioning, which we corrected after scanning the first five patients. We also observed variability in image noise, likely influenced by factors such as dry skin, which can hinder effective subject grounding and limit noise reduction. Notably, older patients—who are more likely to present with drier skin[12]—seemed particularly affected, underscoring the need for tailored strategies to optimize signal quality in this population. Further technical refinement is ongoing, including the implementation of accelerated imaging protocols to shorten scan time, AI-based denoising and improvements to the grounding sleeve design to ensure more consistent image quality.
Beatrice LENA (Leiden, The Netherlands), Simonetta R.g. VAN GRIETHUYSEN, Dennis A. TON, Berend C. STOEL, Denis P. SHAMONIN, Javad PARSA, Ruben B. VAN DEN BROEK, Chloé F. NAJAC, Yiming DONG, Yanli LI, Annette H.m. VAN DER HELM - VAN MIL, Andrew WEBB
|
Auditorium 900 |
|
"Saturday 11 October"
Added to your list of favorites
Deleted from your list of favorites
B30
08:30 - 09:30
FT3-4 - Quality of dissemination in research outputs
FT3: Cycle of Quality
08:30 - 09:30
Publications, working in collaborations.
Nikola STIKOV (Keynote Speaker, Montreal, Canada)
08:30 - 09:30
Quality assessment of published research.
Stefano MOIA (Keynote Speaker, The Netherlands)
|
Espace Vieux-Port |
|
"Saturday 11 October"
Added to your list of favorites
Deleted from your list of favorites
C30
08:30 - 09:30
FT3 Oral - Increasing quality across modalities and organs
08:30 - 08:40
#47883 - PG037 Contrasting the use of whole-body diffusion-weighted MRI and 18F-FDG PET/CT for the metastatic evaluation of paediatric malignancies: a systematic review and meta-analysis.
PG037 Contrasting the use of whole-body diffusion-weighted MRI and 18F-FDG PET/CT for the metastatic evaluation of paediatric malignancies: a systematic review and meta-analysis.
Paediatric cancers represent a small but significant spectrum of childhood and adolescent diseases with a substantial global disease burden. While Positron Emission Tomography/Computed Tomography (PET/CT) is one of the primary imaging modalities used for the staging and metastatic evaluation of paediatric malignancies, its reliance on ionising radiation poses significant risks for paediatric patients, who are highly radiosensitive. Whole-Body Magnetic Resonance Imaging (WB-MRI) is an emerging radiation-free modality that may offer a safer alternative for the detection of metastases in paediatric malignancies; however, its accuracy requires further validation. This systematic review aimed to synthesise the available evidence to comparatively evaluate the use of whole-body diffusion-weighted MRI (WB-DWI) as a radiation-free alternative to PET/CT for the metastatic evaluation of paediatric cancers.
A comprehensive literature search was conducted from September to December 2024 across PubMed, Embase and Web of Science. Inclusion criteria were: participants assessed by both WB-MRI and 18F-fluorodeoxyglucose PET/CT (18F-FDG PET/CT); histopathological confirmation of the primary malignancy was present; and participants aged ≤21. Exclusion criteria were: diffusion-weighted sequencing not used in the WB-MRI protocol; hybridised PET/MRI evaluated; or true positive (TP), false positive (FP), true negative (TN), and false negative (FN) data could not be extracted. Applicability and risk of bias were assessed using the Quality Assessment of Diagnostic Accuracy Studies (QUADAS)-2 tool. Pooled sensitivity, specificity and predictive values were calculated using a univariate weighted analysis. 9 studies met the criteria to be included in this review, and 8 were included in the meta-analysis. Figure 1 displays the PRISMA flow chart representing the selection of studies. The malignancies represented in the meta-analysis were all haematological in nature, primarily assessing Hodgkin lymphoma, with more limited data for Langerhans cell histiocytosis and non-Hodgkin lymphoma. The excluded study assessed patients with neuroblastoma and was analysed separately due to a high risk of bias and methodological incompatibilities. All studies were judged to be at a high or unclear risk of bias in at least one QUADAS-2 domain.
Compared against PET/CT, the pooled per-patient and per-lesion sensitivity values of WB-MRI were 91.7% (95% CI: 77.5-97.3%) and 87.6% (95% CI: 78.0-94.2%), respectively. Using a per-region analysis, sensitivity was 94.9% (95% CI: 90.2-97.7%) and specificity was 98.7% (95% CI: 96.9-99.4%). The pooled sensitivity and specificity values are represented in Figure 2. While overall performance of the two modalities was comparable, some variability was observed, particularly in studies with smaller sample sizes. False negative WB-MRI results occasionally led to under-staging; however, not all discrepancies were clinically significant. WB-MRI showed a high level of agreement with PET/CT for the identification of metastases, particularly for nodal disease. Diagnostic accuracy varied across anatomical locations; however, for overall staging, accuracy was high, with few patients incongruently staged by the two modalities such that there were implications for treatment. While PET/CT formed the reference standard for this review, it should be noted that PET/CT is not a perfect diagnostic test, and so discrepancies between the two modalities are not necessarily always indicative of a WB-MRI error. Without an objective, independent reference – such as histopathological confirmation of any identified lesions, or additional imaging – the true source of discordance between the two modalities cannot be conclusively established. Additionally, despite the substantial advantage that WB-MRI offers by eliminating radiation exposure, there are limitations to the implementation of WB-MRI. Limited availability of MRI scanners and longer scan durations compared to PET/CT increase the likelihood of sedation being required, introducing additional clinical risks. Quantifying the diagnostic value of specific MRI sequences is therefore essential for the development of evidence-based MRI protocols to minimise scanning times. WB-DWI MRI demonstrated a high concordance with 18F-FDG PET/CT, however, rarer malignancies remain underrepresented in the literature and may rely on the generalisability of other results to guide clinical recommendations. Where serial imaging is required and cumulative radiation exposure is a concern, WB-MRI offers an advantage over PET/CT imaging. With further supporting evidence and appropriate protocol optimisation, WB-MRI may offer a viable, radiation-free alternative to PET/CT and may reshape staging practices in paediatric oncology by reducing radiation burden without compromising on diagnostic accuracy.
Leanne MAHER (Lincoln, United Kingdom), Daniel MCLAUGHLIN
08:40 - 08:50
#47739 - PG038 Diffusion MRI preprocessing impacts ADC estimation and automatic PI-RADS v2.1 classification in bi-parametric prostate MRI.
PG038 Diffusion MRI preprocessing impacts ADC estimation and automatic PI-RADS v2.1 classification in bi-parametric prostate MRI.
Prostate cancer (PCa) is the most common type of malignancy in the male population. Bi-parametric Magnetic Resonance Imaging (bp-MRI) combining T2-weighted (T2w) imaging with Diffusion MRI (dMRI), has been proven non-inferior to multi-parametric MRI (mp-MRI) for PCa detection [1]. DMRI-derived Apparent Diffusion Coefficient (ADC) maps are widely used to detect PCa, but dMRI suffers from artifacts that affect the quality of ADC maps. This study aimed to evaluate the impact of dMRI preprocessing on the estimation of ADC maps, with a goal to automatically determine PI-RADS [2],[3] scores based on bp-MRI sequences. As PI-RADS 3 lesions are radiologically further assessed using intravenous contrast, we aimed to automatically classify lesions as PI-RADS 3 or not, to facilitate workflow as the patient lies in the scanner.
Data: 268 cases from fastMRI prostate dataset were used [4], containing T2w and dMRI axial scans with individual-slice PI-RADS labels. Each case contained 48 dMRI images with two different b-values (b=50, 1000 s/mm²). Preprocessing: 5 pipelines were compared: 1) averaged unprocessed, 2) non-averaged unprocessed, 3) non-averaged denoised [5], 4) denoised and Gibbs-ringing corrected [5] and 5) denoised, Gibbs-ringing corrected and susceptibility distortion corrected by registration to the T2w along the phase encoding direction [5]. ADC estimation: for each pipeline iterative weighted linear least squares (IWLLS) [6] and LLS regression were performed, resulting in 10 differently processed ADC maps. Segmentation: axial T2w sequences were used as input for a pretrained, in-house finetuned UNet-based algorithm [7] to obtain segmentations of the prostate. PI-RADS classification: A DenseNet [8] architecture with 16 initial features and a block configuration of [6,12,24,16] was used for 3-class classification, comparing all pipelines with IWLLS estimation. PI-RADS scores 1,2 (class 1) and scores 4,5 (class 3) were grouped together. PI-RADS 3 patients represented class 2. Model input consisted of multi-channel 2D slices including: the T2w image, the IWLLS-estimated ADC map and either the averaged dMRI data for pipeline 1 or the non-averaged dMRI data for the rest. Adam optimizer [9] was used with a learning rate of 1e-5. Training was paused after 230 epochs when no significant improvement in per-class Area Under Receiver Operating Curve (AUROC) was observed. A validation set of [6/4/3] patients per class [1/2/3] was used to compare slicewise model performance, by comparing the average AUROC values per class for each pipeline. Optimal decision thresholds per class were calculated on the validation set for the best performing pipeline using Youden’s Index [10]. Model weights from the last training epoch of the best performing pipeline were applied per slice on a test set of 37 patients (17/11/9). Although Wilcoxon signed-rank tests revealed statistically significant differences across mean slice-wise ADC estimates for all pipelines (p < 0.001), Pearson Correlation Coefficients (PCCs) were consistently high (0.975–1.000) within the prostate mask for all pipeline combinations. Figure 1 shows the differences in an axial IWLLS-estimated ADC slice across pipelines. Figure 2 displays a Bland-Altman plot of the two most different mean slicewise ADC distributions based on their lowest pcc value: pipeline 3 LLS and pipeline 5 IWLLS. Classification results are displayed in table 1. Pipeline 5 (fully processed data) was superior (AUROC values 0.685-0.827), yielding slicewise AUROC values of 0.852, 0.826 and 0.909 for classes 1,2 and 3 respectively on the independent test set. Sensitivity and specificity were reported 0.845/0.725, 0.667/0.841 and 0.316/0.993 for classes 1,2 and 3 respectively. Figure 3 shows the slicewise per-class ROCs. dMRI preprocessing had a significant effect on ADC estimation and was associated with statistically significant differences in mean ADC values across slices. Nevertheless, the high PCC values indicate preserved contrast in dMRI images independently of the preprocessing method. Automatic PI-RADS classification performance, however, was more sensitive to preprocessing. Susceptibility distortion correction led to the largest performance gain, underscoring the importance of accurate dMRI-to-T2w alignment for AI-driven analysis. Additionally, the benefit of using raw dMRI data over scanner-averaged sequences for clinical use was demonstrated. These findings highlight how preprocessing choices critically influence downstream automatic tasks like PI-RADS classification. Overall, results demonstrate that dMRI preprocessing has a dual impact: it alters mean ADC values significantly without disrupting diffusion patterns and, more critically, it enhances the reliability of dMRI data in supporting automated PI-RADS scoring and downstream clinical decision-making. This underscores the importance of selecting appropriate preprocessing strategies for robust AI-driven diagnostics in prostate MRI.
Christos KANAKIS (Utrecht, The Netherlands), Mathias PERSLEV, Tim SCHAKEL, Silvia INGALA, Michael Bachmann NIELSEN, Akshay PAI, Dennis W.j. KLOMP, Chantal M.w TAX
08:50 - 09:00
#47576 - PG039 Setting up a standardised Whole Body MRI clinical protocol for patients with myeloma across 10 UK hospitals.
PG039 Setting up a standardised Whole Body MRI clinical protocol for patients with myeloma across 10 UK hospitals.
Whole Body (WB) MRI is embedded in national [1] and international [2] guidance for myeloma imaging. However, a national survey data suggests that protocol optimisation and radiographer training are barriers to implementation of such a service [3]. This initiative aimed to deliver radiographer training and a standardised clinical WB-MRI protocol for early diagnosis of myeloma across 10 sites within the RM Partners West London Cancer alliance, UK.
Figure 1 summarises the project steps and participating scanners. All sites were visited during an eight-month period (October 2024 - May 2025). The standardised protocol derived from a 1.5T Sola Siemens scanner and was compliant with the MY-RADS core protocol guidelines [4]. The protocol includes multi-station axial acquisitions of Diffusion Weighted Imaging (DWI) and Dixon sequences; seven stations were utilised to cover an average adult from vertex to knees.
The site visit (including a physicist and radiographer) supported protocol set up and provided practical training on healthy volunteer data acquisition and scanning of a DWI phantom. DWI acquisition of a single station from the WB-DWI protocol was performed using a calibrated phantom (Diffusion Phantom Model 128 [serial number 128-0230], Caliber MRI, Boulder, Co. USA, [5]). The same phantom was scanned in the same holder and orientation on each scanner. The Apparent Diffusion Coefficient (ADC) measurements were compared with the tabulated room-temperature-specific values available from the phantom manufacturer.
Details of the MR protocol utilised for each vendor are presented in Figure 2. In order to support clinical sites with MRI capacity challenges, the WB-MRI protocol was optimised to accelerate the DWI acquisition (i.e. using a 2 b-value sequence and, where available, the artificial intelligence (AI)-based DWI reconstruction). The maximum DWI acquisition time across all seven stations was ~30 min.
The site qualification process included review of the volunteer data and quantitative assessment of the DWI phantom. The standardised WB-MRI protocol was successfully set up at 10 UK scanners/sites involving three vendor systems. The ADC measurements across all scanners (Figure 3) showed a relative ADC measurement error of < 10% across a wide clinical range of ADCs (e.g. 50-200 x 10-5 mm2/s). A relatively large room temperature range was observed across the 10 scanners (18 - 23 °C).
Exemplar images from three healthy volunteers are presented in Figure 4, showing axial b900, ADC and fat-only Dixon images of the pelvis station, together with maximum intensity projection (MIP) of the coronal reconstruction of multi-station axial b900 images. Various hardware and software limitations were observed. Two sites had lower specification gradients limiting the maximum gradient strength (e.g. 33 mT/m instead of 45 mT/m) and slew rate (125 T/m/s instead of 200 T/m/s) of the diffusion gradients, thus resulting in longer echo times and consequently slightly longer acquisition times. The GE and Philips scanners in this evaluation did not offer tools on the console/workstation for image arithmetic to calculate relative fat fraction [FF=F/(F+W)] from the T1w Dixon imaging. Across these 10 scanners, only the GE sites had access to AI-based DWI reconstruction for acceleration of DWI at the time of this study.
Nevertheless, whilst accepting minimal site-specific modifications of the optimised MR sequence parameters, we were able to share the methodology and imaging protocols resulting in images which were suitable for qualitative and quantitative analysis, with accurate ADC estimates obtained in phantom measurements at all 10 sites. Patient data acquisition is ongoing and potential slight changes of the protocol may still occur. Standardised WB-MRI protocols can be implemented and supported across the three main vendor systems allowing all hospital sites in the network to scan myeloma patients. With minimal modification, this protocol can also be applied to emerging WB-MRI applications in metastatic bone disease and screening high risk populations.
Mihaela RATA (London, United Kingdom), Alison MACDONALD, Georgina HOPKINSON, Richard NORCLIFFE, Lesley-Anne HAMMOND, Edith BENDANA, Rina KAPADIA, Pablo MARTINPOLANCO, Rosaleen O'KEEFFE, Aman JUTTLA, Jamal SALEH, Olga FADEEVA DA COSTA, Tara BARWICK, Jimaio UBALDO, Jessica WINFIELD, Christina MESSIOU
09:00 - 09:10
#45950 - PG040 Normative Diffusion MRI Metrics of the Cervical Spinal Cord Across Age Groups on a 3T Hybrid PET/MRI Scanner.
PG040 Normative Diffusion MRI Metrics of the Cervical Spinal Cord Across Age Groups on a 3T Hybrid PET/MRI Scanner.
Normative MRI datasets of the spinal cord are essential for detecting deviations linked to neurological disorders. However, age-specific reference values remain scarce, limiting their application in clinical settings. This study aims to characterize age-related microstructural changes in the cervical spinal cord using diffusion MRI. We focused on the gracilis, cuneatus, and lateral corticospinal tracts due to their known involvement in multiple sclerosis (MS) [1], to support future biomarker development. Notably, this is the first normative dataset acquired with a custom protocol developed for the Siemens Biograph mMR hybrid PET/MRI scanner, a system not previously optimized for spinal cord imaging.
Thirty-six healthy individuals were scanned on a 3T Siemens Biograph mMR hybrid PET/MRI system using a dedicated protocol including T2-weighted and DTI sequences. The cohort comprised 17 young (mean age = 30.4 ± 4.1 years, 13 female) and 19 elderly subjects (mean age = 60.8 ± 5.1 years, 13 female).
T2-weighted images were acquired using a 3D turbo spin echo sequence (TR/TE = 1500/120 ms, flip angle =120°, voxel size = 0.8 × 0.8 × 0.8 mm³). DTI was performed according to international guidelines for quantitative MRI of the spinal cord, using a spin-echo EPI sequence (TR/TE = 670/76 ms, 30 directions, b = 800 s/mm², 5 b0 volumes, voxel size = 0.9 × 0.9 × 5 mm³) [2].
Images were processed using the Spinal Cord Toolbox (SCT) for segmentation and vertebral labeling (C1–C6) [3]. Diffusion metrics—FA, MD, AD, RD—were extracted from the gracilis, cuneatus, and lateral corticospinal tracts bilaterally. Statistical comparisons were performed using two-sample t-tests and Cohen’s d, with FDR correction. Results were visualized with bar and volcano plots. FA and AD showed the most consistent age-related differences. The right gracilis tract at C2 exhibited the strongest effect (p = 0.0012, d = 1.17), remaining significant after FDR correction. Other features with large effect sizes (d > 0.8), although not surviving correction, were observed in the lateral corticospinal and gracilis tracts for MD, AD, and RD.
Figure 1 shows mean ± SD of all metrics across tracts and vertebral levels (C1–C6). Elderly subjects had lower FA and higher RD values. The right lateral corticospinal tract at C2–C3 showed the highest inter-subject variability in diffusivity.
Figure 2 presents line plots highlighting higher FA and lower RD in young participants, especially in posterior and lateral tracts.
Figure 3 combines volcano plots for all four metrics. Only FA in the right gracilis at C2 remained significant after FDR correction. Several features fell in the large-effect zone (|d| > 0.8), especially in lateral corticospinal tracts for AD and MD (Figure 3). This is the first normative diffusion MRI dataset of the cervical spinal cord acquired on a hybrid PET/MRI system. By combining tract-specific analysis with quantitative diffusion metrics, we identified robust and anatomically consistent age-related differences. Reductions in FA and increases in RD and MD, particularly in sensorimotor pathways, align with expected aging-related demyelination and axonal loss [4,5].
Methodologically, we provide standardized diffusion profiling across C1–C6, using a reproducible pipeline [3]. The integration of high-resolution anatomical and diffusion imaging within a PET/MRI platform enhances translational applicability.
Identified features with high effect sizes and low variability offer a basis for biomarkers in MS and may aid in early diagnosis and monitoring [1]. This normative framework also enables future investigations into comorbidities and sex differences in spinal cord aging [6]. We present a tract- and level-specific normative dataset of cervical spinal cord diffusion metrics across age groups, acquired using a custom protocol on a hybrid PET/MRI system. Results confirm robust and anatomically localized microstructural changes with aging, particularly in MS-relevant tracts. This dataset provides a reference framework for precision MRI biomarkers in neurodegenerative and inflammatory disorders.
This work was supported by the D³4Health initiative (Project PNC0000001), funded by the Italian Ministry of University and Research under the National Plan for Complementary Investments to the PNRR (PNC-PNRR).
Alessia SARICA (Catanzaro, Italy), Maria Eugenia CALIGIURI, Chiara CAMASTRA, Ilaria CHIMENTO, Paola VALENTINO, Stefania BARONE, Umberto SABATINI, Andrea QUATTRONE, Aldo QUATTRONE
09:10 - 09:20
#47845 - PG041 Breast tissue segmentation at 7T using Chemical Exchange Saturation Transfer (CEST).
PG041 Breast tissue segmentation at 7T using Chemical Exchange Saturation Transfer (CEST).
Breast tissue composes mostly of fibroglandular tissue (FGT) and fat, and the differentiation of these tissues is key for breast cancer diagnosis [1]. While CEST MRI is mainly used to probe metabolites, its frequency-selective saturation enables water-fat separation [2]. Beyond this, multidimensional CEST data may support more detailed breast tissue segmentation. We explore dimensionality reduction and clustering of Z-spectra for this purpose.
A CEST MRI sequence was implemented using 120 Gaussian-shaped saturation pulses (15.360 ms, 10 ms interpulse delay, B1 = 1 μT). Imaging readout used a snapshot acquisition (TR: 7.4 ms, TE: 1.46 ms, FA: 6°) [3]. 41 frequency offsets (0.25 ppm steps) were acquired from -5 to +5 ppm with 1 s recovery. All measurements were performed on a 7T MAGNETOM Terra.X scanner (Siemens Healthcare, Erlangen) using a local Rapid 4Tx/16Rx 1H breast coil, in three healthy female volunteers, with ethics approval and informed consent. Although a 4Tx system, the coil ran in fixed protected mode. For Volunteer 1, B1 was 1 μT; for Volunteers 2 and 3, reduced to 0.8 μT to meet SAR limits.
Simulated CEST spectra were created with the BMCTool package [4]. For segmentation, dimensionality reduction (PCA, UMAP) [5,6] was applied to voxel-wise spectral data, followed by k-means and agglomerative clustering [7-8]. FGT segmentation was evaluated by computing the Dice Similarity Coefficient (DSC) against a reference manually segmented by a trained radiologist based on 1 CEST image and a DIXON acquisition. Simulated Z-spectra of voxels containing only water, only fat, and a 50/50 mix show the influence of fat with a second dip at -3.5 ppm, the resonance frequency assumed in this work for the main fat peak (Figure 1a). Experimental spectra under matching conditions show similar features (Figure 1b). Figures 1(c-f) display CEST images at 4 saturation offsets, illustrating natural suppression of fat or water at different frequencies.
In Figure 2a, the PCA component loading vectors are shown, with Figures 2(c-f) displaying the corresponding images. The second principal component (Fig. 2d) reveals a clear fat-water contrast, reflected in the loading vector through opposite weighting of fat and water frequencies. Figure 2b presents the PCA Cumulative Explained Variance, indicating 3 components explain 95% of the data and 6 explain 98%, with the curve stabilizing around 20 components. In different regions of interest (ROI), the cumulative explained variance does not hold.
Figure 3 presents results from two clustering algorithms (k-means, agglomerative) using the full dataset, 3 and 21 PCA components, and 3- and 20-dimension UMAP reductions. Figures 3a and 3b show a raw CEST image and an FGT mask manually delineated by a radiologist on the image in 3a.
In the “whole FOV” results, both raw and PCA data consistently subdivide FGT into two clusters: one at the periphery and one in the interior, likely due to partial volume effects. Only 20-dimension UMAP placed all FGT voxels into a single cluster, yielding the highest DSC for FGT. However, 3-dimension UMAP (Fig. 3(o-p)) resulted in good segmentation of the pectoral muscle, though not with complete efficacy as the muscle shares a cluster with some FGT voxels. Generally, both fat and FGT split into several clusters, leaving uncertainty whether these should be combined into a single mask or if subdivisions reflect meaningful variation.
To explore CEST’s potential for differentiating FGT into ducts and glands, clustering was applied to a mask containing only FGT tissue (“Only fibroglandular tissue” column). Distinct localized clusters emerged, though no suitable reference is available to confirm anatomical correspondence. The most striking result appears in 3v, where 3 similar clusters (9, 10 and 11) are located near the nipple region, where higher duct density is expected. However other reasons (anatomical and technical) might be influencing this observation.
Figure 4 further examines the results from the 20-dimension UMAP by showing the Z-spectra (4b, d) for ROIs of each cluster from the “Whole FOV” and “Only Fibroglandular Tissue” analyses. The clustering differentiates regions with higher noise, greater fat contribution, and stronger water peaks. In particular, Figure 4d highlights varying fat peak amplitudes, suggesting clustering reflects fat fraction variations. This work demonstrates that CEST’s spectral data structure enables tissue clustering beyond fat-water separation. While fat and fibroglandular tissue often subdivide, this may reflect not only partial volume effects and field inhomogeneities but also additional contrast from CEST. Notably, UMAP-reduced data yielded the most coherent FGT segmentation. If the goal is purely fat-FGT separation, thresholding CEST images at -3.5 ppm or using the 2nd PCA component might suffice. However, the observed localized clusters suggest potential for meaningful tissue differentiation, warranting anatomical validation.
Magda DUARTE (Erlangen, Germany), Katharina BREININGER, Katharina TKOTZ, Sebastian BICKELHAUPT, Moritz ZAIß
09:20 - 09:30
#47665 - PG042 A novel deep learning pipeline for 17-segment myocardial scar segmentation and quantification in CMRI.
PG042 A novel deep learning pipeline for 17-segment myocardial scar segmentation and quantification in CMRI.
Accurate myocardial scar segmentation is vital for stratifying sudden cardiac death risk, ventricular arrhythmias, prognosis, and therapy planning [1]. Late gadolinium enhancement (LGE) CMR assesses left ventricular (LV) scar tissue, aiding arrhythmia risk evaluation [2]. However, automated segmentation is difficult due to subtle scars [3] and anatomical variability [4]. The AHA 17-segment model standardizes LV division across imaging modalities for consistent analysis [5]. Its coronary alignment enables precise ischemia localization, and automation may boost diagnostic accuracy [6]. Yet, current methods often need predefined LV contours and manual refinement [7], highlighting the need for a fully automated deep learning (DL) framework.
We propose a тщмуд DL pipeline for automated LV scar segmentation using the 17-segment model. It includes: (1) DL-based slice classification and landmarks detection, (2) segmentation of healthy myocardium and scar, (3) algorithmic 17-segment partitioning. This approach accelerates processing, improves accuracy, reduces manual input, and supports clinical decision-making.
Pipeline overview. The DL-assisted pipeline (Figure 1) performs automated 17-segment myocardial scar segmentation and quantification. It starts with a ResNet50 model that classifies MRI slice levels, identifies the LV center of mass (C), and detects an anatomical reference point (P) (uppermost point of the attachment of the right ventricular wall to the left ventricle). LV center of mass guides image cropping. The cropped images are then processed by a U-Net model to segment healthy myocardium and scar. Outputs from both models feed into a mathematical algorithm that divides the LV into 17 segments following AHA guidelines. The multi-segment mask is combined with the scar mask to localize and quantify scar volume in each of the 17 segments. A bull’s-eye plot then visualizes scar distribution for intuitive interpretation.
Dataset. The MRI dataset includes data of 150 post-infarction cases from ANMRC (130 for training/validation, 20 for testing) and 100 labeled cases from the EMIDEC dataset [8], both with delayed contrast-enhanced T1-weighted images for scar detection. ANMRC images were manually segmented in MedSeg by a medical physicist under radiologist supervision; EMIDEC provided annotated masks. Each slice was manually classified into four anatomical levels, anatomical reference points were identified, and 17-segment masks were generated semi-automatically following AHA standards [9]. Ground truth scar volume per segment was derived from these annotations.
DL Models. Two DL models were used: ResNet50, with classification and regression output layers, for slice classification and anatomical landmarks prediction and U-Net (enhanced with attention mechanisms) for healthy myocardium and scar segmentation. To improve accuracy, images were cropped around the LV centroid and used as inputs for the U-Net model.
Segmentation performance was evaluated using Dice Similarity Coefficient (DSC), anatomical landmarks (C and P ) prediction with mean absolute error (MAE), while scar volume estimation with both MAE and Pearson correlation. The ResNet50 model achieved 86% slice classification accuracy, with MAE of 2.60 and 4.30 pixels for the LV centroid (C) and reference point (P), respectively. U-Net showed strong performance with median DSCs of 0.84 for healthy myocardium and 0.74 for scar segmentation (Figure 2a). The trained networks were integrated into the pipeline. A mathematical algorithm based on their outputs achieved a mean DSC of 0.85 ± 0.05 across the 17 segments (Figure 2b–c). Scar quantification showed a 7.63% MAE and a Pearson correlation of 0.86, indicating strong agreement with the ground truth. Our deep learning-assisted pipeline for myocardial scar segmentation within the AHA 17-segment model demonstrates competitive performance compared to existing methods. While the scar quantification error (MAE 7.63%) is slightly higher than a previous semi-automatic approach (6.4%) [9], the proposed method is fully automated, enhancing processing speed. Additionally, the 17-segment myocardial segmentation achieved a higher mean DSC (0.85 ± 0.05) than a recent automated method (DSC 0.81) reported in [7], highlighting its strong potential for clinical application. The developed pipeline was implemented as a desktop application with a simple and user-friendly interface, as shown in Figure 3. The proposed deep learning-assisted pipeline offers a fully automated and accurate solution for myocardial scar segmentation using the AHA 17-segment model. By integrating deep learning with mathematical modeling, it streamlines analysis, reduces manual effort, and supports standardized clinical interpretation. Future work will aim to improve performance in apical regions and validate the method across larger, diverse datasets.
This study was supported by the Russian Science Foundation (RSF) grant No. 23-75-10045
Walid AL-HAIDRI (Saint Petersburg, Russia), Levchuk ANATOLIY, Kseniya BELOUSOVA, Vladimir FOKIN, David BENDAHAN, Ekaterina BRUI
|
Salle Major |
|
"Saturday 11 October"
Added to your list of favorites
Deleted from your list of favorites
D30
08:30 - 09:30
FT2-1 - Imaging early life
FT2: Cycle of Translation
08:30 - 09:30
Brain MRI on awake pediatric patients.
Stefan SKARE (Keynote Speaker, Sweden)
08:30 - 09:30
MRI of fetal neurodevelopment.
Daniela PRAYER (Keynote Speaker, Vienna, Austria)
|
Salle 120 |
|
"Saturday 11 October"
Added to your list of favorites
Deleted from your list of favorites
E30
08:30 - 09:30
EFRS
|
Salle 76 |
09:30 |
TIME FOR A BREAK
- Coffee and refreshments will be available at the cash bar.
|
09:50 |
"Saturday 11 October"
Added to your list of favorites
Deleted from your list of favorites
A31
09:50 - 10:50
FT1 Plenary
The renascence of gradients for high performance MRI
FT1: Cycle of Technology
09:50 - 10:50
Diffusion imaging with ultra-strong gradients across the body.
Chantal TAX (Associate Professor) (Keynote Speaker, Utrecht, The Netherlands)
09:50 - 10:50
Pushing the limits of high-performance gradients: design and applications.
Markus WEIGER (PhD) (Keynote Speaker, Zurich, Switzerland)
|
Auditorium 900 |
10:50 |
TIME FOR A BREAK
- Coffee and refreshments will be available at the cash bar.
|
11:00 |
"Saturday 11 October"
Added to your list of favorites
Deleted from your list of favorites
A32
11:00 - 12:30
FT1-2 - RF transmission and reception
Advanced techniques and applications
FT1: Cycle of Technology
11:00 - 12:30
Metamaterials in MRI or X-nuclei imaging.
Rita SCHMIDT (Senior Scientist) (Keynote Speaker, Rehovot, Israel)
11:00 - 12:30
pTx in practice.
Christoph AIGNER (Research Scientist) (Keynote Speaker, Berlin, Germany)
11:00 - 12:30
SAR management in pTx.
Andreas BITZ (Keynote Speaker, Aachen, Germany)
11:00 - 12:30
Tailor-made: Wearable RF coils (Flexible, wireless).
Martijn CLOOS (PhD) (Keynote Speaker, Nijmegen, The Netherlands)
|
Auditorium 900 |
|
"Saturday 11 October"
Added to your list of favorites
Deleted from your list of favorites
B32
11:00 - 12:30
ET1-2 - Data visualization and figure creation
11:00 - 12:30
Coloring your MR maps.
Miha FUDERER (Keynote Speaker, Utrecht, The Netherlands)
11:00 - 12:30
Interactive brain plots.
Saige RUTHERFORD (Keynote Speaker, Jena, DE, The Netherlands)
11:00 - 12:30
Interactive scatter plots.
Saige RUTHERFORD (Keynote Speaker, Jena, DE, The Netherlands)
|
Espace Vieux-Port |
|
"Saturday 11 October"
Added to your list of favorites
Deleted from your list of favorites
C32
11:00 - 12:30
FT3 LT - Increasing quality in AI applications
11:00 - 11:02
#47923 - PG205 Enhancing Hippocampal Subfield Visibility from Repeated 3T MRI: High-SNR Image Generation for Deep Learning-Based Segmentation.
PG205 Enhancing Hippocampal Subfield Visibility from Repeated 3T MRI: High-SNR Image Generation for Deep Learning-Based Segmentation.
Accurate segmentation of the hippocampus and its subfields is critical for studying neurodegenerative diseases such as Alzheimer’s. These submillimeter structures require high-resolution MR images with strong signal- and contrast-to-noise ratios (SNR, CNR). These are difficult to achieve at 3T due to the hippocampus’s small size and deep brain location.
A common approach for imaging the hippocampus is to use 2D T2-weighted (T2w) turbo-spin-echo (TSE) sequences, with very high in-plane resolutions and comparably thick slices. However, low SNRs and CNRs often compromise visibility of the fine internal hippocampal architecture. To address these limitations, within-subject averaging of multiple repeated acquisitions has been proposed to enhance image quality [1, 2]. When combined with robust preprocessing, such as N4 bias field correction and non-linear registration, this strategy can significantly boost anatomical fidelity.
Here, we compare the generation of a high-SNR image using 25 repeated 3T T2w scans from two healthy subjects, aligned via a SyN-based ANTs pipeline. We compare simple averaging with a gradient-based method emphasizing structural boundaries, aiming to support improved visualization and ground truth annotation for deep learning-based hippocampal subfield segmentation.
Dataset.
We acquired a unique dataset consisting of 25 repetitive T2w scans (3T Siemens Prismafit) of two healthy subjects. T2w scans were obtained using a 2D fast spin-echo with hyperechoes (48 slices, 0.47×0.47×1 mm³; 352×512×48 matrix) in an oblique coronal plane perpendicular to the long axes of the hippocampi.
Image alignment.
All T2-weighted volumes were aligned using a multi-stage registration pipeline implemented with the Advanced Normalization Tools (ANTs) software suite [3,4]. We applied symmetric normalization (SyN), a diffeomorphic model that generates smooth, invertible deformation fields suitable for high-resolution MRI registration [5].
To account for local intensity variations, common in T2-weighted imaging, the Mattes mutual information metric was used [6]. A four-level multi-resolution strategy (100, 70, 50, 20 iterations) with 32% voxel sampling per level was used to balance accuracy and efficiency [4,7].
The reference volume was selected based on the lowest Frobenius norm of intra-volume transformations between odd and even slices, indicating minimal inter-slice distortion and highest internal consistency. Prior to registration, all scans were N4 bias-corrected [8], padded by 20 mm in each spatial direction to reduce edge effects, and histogram-matched to the reference.
Image Combination / Averaging.
Following spatial alignment, which ensured a reliable basis for the subsequent voxel-wise high-SNR reconstruction, an average and gradient-based fusion approach was employed to synthesize a high-SNR T2-weighted volume from the aligned inputs. This method enhanced the representation of anatomical structures by prioritizing edges and tissue boundaries, which typically carry greater structural information.
In particular, local gradient magnitudes were computed for each volume using the Gaussian gradient magnitude operator with a fixed standard deviation (σ = 1). Calculated gradients were then used to create weight maps. This process emphasizes smooth intensity transitions and edges while suppressing noise [9,10]. The final high-SNR volume was obtained by computing the weighted average voxel intensities. For comparison, also simple voxel-wise averaging of the aligned images was calculated. To assess the quality of the resulting high-SNR images, SNR values were computed for the reference images and their corresponding batch-averaged reconstructions (N = 9, 16, 25). Quantitatively, the SNR of Subject 1 increased from 42.27 (original) to 59.99 using Gaussian-weighted averaging with a batch size of 25. For Subject 2, SNR improved from 35.56 to 54.44 under the same conditions. The complete set of SNR measurements across methods and batch sizes is summarized in Table 1. A qualitative comparison of the resulting images is shown in Figures 1 to 3. Quantitative and qualitative assessments demonstrated consistent improvements in SNR across all reconstruction methods compared to the original T2-weighted MR images. Visual comparison (cp. Fig. 1 to Fig. 3) reveals enhanced anatomical detail and reduced noise in images reconstructed with Gaussian gradient weighting, particularly at higher batch sizes. Comparison of SNR values (Tab. 1) confirms that Gaussian weighting consistently outperforms simple averaging across all tested batch sizes. This study demonstrates the benefit of generating high-SNR T2-weighted hippocampal images through averaging of repeated 3T acquisitions, with gradient-based Gaussian weighting consistently outperforming simple averaging. Resulting images offer improved anatomical fidelity, supporting more reliable downstream applications such as ground truth data for deep learning-based segmentation.
Maximilian SACKL (Graz, Austria), Marlene SCHICHL, Stefan ROPELE
11:02 - 11:04
#45793 - PG206 From Proof-of-Concept to Clinical Validation: Robust Automated Segmentation of Diffuse Lower-Grade Gliomas for Longitudinal Monitoring.
PG206 From Proof-of-Concept to Clinical Validation: Robust Automated Segmentation of Diffuse Lower-Grade Gliomas for Longitudinal Monitoring.
The monitoring diffuse lower-grade gliomas (DLGG) presents significant challenges due to the tumor's infiltrative nature and post-surgical brain remodeling, complicating MRI assessment. Traditionally, 2D tumor size measurements, as recommended by RANO criteria, have been used. Yet, they fail to capture subtle volumetric changes that are critical for tracking tumor progression. Volumetric assessment offers a more accurate estimation of tumor behavior but has so far been dependent on time consuming manual segmentation, rendering it impractical in routine clinical practice. Our previous proof-of-concept study demonstrated the feasibility of a nnU-Net model for DLGG segmentation, but clinical validation with larger, more heterogeneous datasets is essential. Indeed, existing literature reports encouraging results in low-grade glioma segmentation, but mainly on preoperative data, without incorporation longitudinal or post-surgical data.
This study was based on a cohort of 207 DLGG patients and 1971 MRI exams (Table 1) with longitudinal follow-up (9.55 exams ± 8.52). MRI imaging came from different MRI scanners (Siemens/GE/Philips) at different field strengths (1.5T/3T), including both 2D and 3D FLAIR acquisitions. The dataset, consisting of T1w and T2FLAIR (2D or 3D) data, was divided into a derivation set (N=1771) for model training and a validation set (N=200) for performance testing, while controlling for the 2D/3D FLAIR ratio in both sets (87.5%/12.5%). The nnU-Net model was trained using a 5-fold cross-validation approach.
We evaluated the model's performance by comparing automated segmentations (AS) with manual segmentations (MS) performed by expert neuroradiologists. The primary evaluation metrics were the Dice Similarity Coefficient (DSC) and Intersection over Union (IoU), which measure segmentation overlap. Secondly, we derived tumor volume and mean tumor diameter (MTD) and compared AS with MS using Lin’s concordance correlation coefficient (CCC) and Bland-Altman tests. To further assess the model’s robustness, we analyzed its performance improvement across multiple training sets, gradually increasing the number of exams from 318 to 1971. The nnU-Net model achieved a median DSC of 0.93 across both derivation and validation sets, with an IoU of 0.86. In the validation set, 64% of the cases showed a very good agreement (DSC ≥ 0.9) between AS and MS, and 31% had a good agreement (DSC between 0.7 and 0.9). Only 5% of cases showed unsatisfactory or poor results (Figure 1). Larger tumors were associated with higher DSC values (p<0.001). Tumor volume and MTD derived from AS showed near-perfect concordance with MS, with CCC values of 0.991 and 0.989, respectively for the validation cohort (Figure 2 and 3). Bland-Altman analysis showed a small underestimation by AS compared to MS, averaging 0.9 cm³ for tumor volume and 0.5 mm for MTD.
The model’s performance improved as the number of training examples increased. With a smaller training set of 318 exams, the DSC was 0.82. Increasing the training data to 1009 exams improved the DSC to 0.89, and with 1771 exams, the DSC reached 0.93. However, adding more data beyond 1771 exams (up to 1971) did not yield further significant performance gains (p=0.84), indicating a plateau in the learning curve. Our findings demonstrate that the nnU-Net model is a robust tool for automated DLGG segmentation, achieving a high level of accuracy that is comparable to manual expert segmentations. The model's performance, particularly in handling both 2D and 3D FLAIR images, shows its flexibility in real-world clinical settings. Importantly, the inclusion of longitudinal follow-up data and post-surgical cases with cavities makes this study more comprehensive compared to other studies, which focused solely on preoperative datasets. This broader scope allows for more accurate monitoring of DLGG progression over time and facilitates integration into clinical workflows. The minor underestimations observed in volume and MTD are unlikely to affect clinical decisions, suggesting that the model can be reliably used for patient follow-up.
Furthermore, our analysis of the model’s performance across varying training set sizes confirms the importance of large, diverse datasets for improving deep learning model accuracy. The plateau observed in performance at 1771 exams suggests that the model has been sufficiently trained for this clinical context, although further external validation on datasets from different centers is recommended. By significantly reducing the time required for segmentation while maintaining high accuracy, this model can enhance clinicians' ability to monitor tumor progression and assess treatment response in longitudinal follow-up. Future research should focus on external validation across diverse clinical environments to ensure generalizability and explore the model’s potential to provide more advanced clinical metrics, such as velocity of tumor expansion.
Jeremy DEVERDUN (Montpellier), Guillaume CLAIN, Margaux VERDIER, Hugues DUFFAU, Nicolas MENJOT DE CHAMPFLEUR, Justine MERIADEC, Mathilde CARRIERE, Amelie DARLIX, Emmanuelle LE BARS
11:04 - 11:06
#46663 - PG207 Implicit neural representations for white matter microstructure parameter estimation with gradient non-uniformity correction.
PG207 Implicit neural representations for white matter microstructure parameter estimation with gradient non-uniformity correction.
The Standard Model (SM) of white matter [1] describes the diffusion MRI (dMRI) signal as arising from the convolution of a fiber orientation distribution function (fODF) with a kernel comprising intra-axonal (sticks) and extra-axonal (zeppelin) water signal contributions. Fitting the SM to noisy dMRI data is challenging with its high-dimensional parameter space, potentially leading to low accuracy, precision, and degeneracy of estimates. Supervised deep learning has shown promise for fitting the SM to in vivo dMRI data [2-5], but it has potential drawbacks such as heavily relying on the choice of training data [6] and retraining for each acquisition scheme. Here, we implement Implicit Neural Representations (INRs) [7-9] for SM parameter estimation. INRs leverage spatial correlations across the brain to produce a continuous representation, in contrast to other methods that fit at voxel level. Furthermore, INRs are unsupervised, noise-robust, can be upsampled to higher resolutions, and can effectively include gradient non-uniformity correction in the fitting process. In this work, the INR method is compared against other machine learning methods and Nonlinear Least Squares (NLLS) fitting.
Ground Truth Generation: One MGH Connectome Dataset (subject 11) [10] was used for generating a ground truth. Acquisitions with Δ 19ms were selected. The SM was fitted with the SMI toolbox [2] to generate a set of realistic SM parameters for axon signal fraction f, intra-axonal diffusivity Di, extra-axonal parallel diffusivity De , extra-axonal perpendicular diffusivity Dp. An anisotropic diffusion filter was applied to generate spatially smooth ground truth maps. The Spherical Harmonics (SH) coefficients plm of the FOD were calculated using Multi-Shell Multi-Tissue Constrained Spherical Deconvolution (MSMT CSD) [11]. The simulated signals corresponding to these parameters were calculated from the SM signal equation with an optimized acquisition protocol [2]: b-values [0, 1000, 2000, 8000, 5000, 2000]mm/s2 , number of directions [6, 20, 40, 40, 35, 15] and B-tensor shape [1, 1, 1, 1, 0.8, 0]. A maximum SH order of lmax = 2 was used and Gaussian noise was added (SNR 50). A signal mask was applied based on white matter segmentation [12].
Fitting methods: The SM was implemented as INR (1) in Pytorch (Figure 1) and compared to three other SM fitting methods: 2) supervised machine learning using the SMI toolbox [2] with standard settings; 3) supervised deep learning as implemented in [3] trained on uniformly distributed parameters, 5e5 samples, 75%/25% training/validation split, Gaussian noise with SNR=50; 4) NLLS implemented in the MATLAB optimization toolbox using Levenberg-Marquardt algorithm . Method 1 was trained on a NVIDIA Titan X GPU for 150 epochs using a mean-squared-error loss. Pearson correlation coefficient ρ and Root-Mean-Squared-Error (RMSE) were used for evaluation.
Gradient non-uniformity correction: One healthy volunteer was scanned on a 3T, 300 mT/m Connectom scanner (Siemens Healthineers) with b-values up to 4000 mm/s2 and B-tensor shape [1, 0.5, -0.5, 0]. SM parameter estimation was performed with the INR without and with correction for gradient-nonuniformities. In the second approach, spatially varying, scanner-specific gradient deviations were used to compute a B-tensor for every voxel [13]. This corrected B-tensor was then used in the forward signal prediction (Figure 1c). Fig. 2a shows scatter density plots of the SM parameters for fitting approaches 1 to 4 . RMSE and ρ show superior performance of the INR model for all parameters, most significant for Di and De . Fig. 2b presents the resulting parameter maps across the brain, with INR representing the white matter structure smoothly, while the other methods show spatial irregularity. Fig. 3 highlights the impact of incorporating gradient non-uniformity correction on parameter estimation, with a notable reduction in De values observed when the correction is applied. Training on the simulated data took 308 seconds, whereas training with gradient non-uniformity correction required 534 seconds. Validation on simulated data demonstrated that the INR method more effectively mitigates noise by leveraging spatial correlations compared to voxel-wise fitting methods.
Gradient non-uniformity correction is cumbersome in supervised machine learning as training data would have to be generated capturing all B-tensor variations. INRs offer straightforward gradient-nonuniformity correction while being independent from training data. The INR method shows reasonable interference times, much faster than NLLS. INR-based approach enables accurate and robust estimation of white matter microstructure, outperforming traditional methods by leveraging spatial continuity, incorporating gradient non-uniformity correction, and eliminating the need for training data.
Gerrit ARENDS (Utrecht, The Netherlands), Tom HENDRIKS, Dennis KLOMP, Edwin VERSTEEG, Anna VILANOVA, Maxime CHAMBERLAND, Chantal TAX
11:06 - 11:08
#47539 - PG208 TAsk-DRiven Experimental Design for Protocol Optimization of Ultra-high Gradient Strength Diffusion-weighted MRI Measurements.
PG208 TAsk-DRiven Experimental Design for Protocol Optimization of Ultra-high Gradient Strength Diffusion-weighted MRI Measurements.
Optimizing diffusion MRI (dMRI) acquisition protocols in clinical settings is essential for improving image quality, reducing scan times, and enhancing sensitivity to tissue microstructure. Traditional optimization methods have mainly focused on minimizing the Cramer-Rao Lower Bound (CRLB)[1], optimizing angular coverage of b-shells[2], or using data interpolation[3] to improve model parameter estimates during MR acquisition design.
However, machine learning and deep learning methods, such as TADRED (Task-Driven Experimental Design)[4], or physics-informed networks[5] can generalize more flexibly to different dMRI models. TADRED is a deep learning framework that combines a subsampling network, which identifies the most informative measurements, with a task network that performs the task. This subsampling process efficiently identifies the most informative subsets of data while training a high-performing task-specific model. TADRED optimizes acquisition protocols by progressively reducing the sample set, enhancing overall efficiency.
In this study, we demonstrate the TADRED-based subsampling approach against a naïve subsampling approach that aims to maximise angular coverage within each shell.
Data Acquisition, Processing and Fitting:
Multi-shell dMRI data[6] were acquired on an ultra-strong gradient system (TE = 80 ms, TR = 5 s) with 1.8 mm slice thickness, 66 slices, and a 120 × 120 mm² FOV. The dataset included seven b-shells up to b = 6 ms/μm² and 266 directions (13 b=0). Gradient spacing (Δ) and duration (δ) were 24 ms and 7 ms. Direction counts scaled with b-values, with interleaved b=0 images to aid correction. Gradient spacing (Δ) and duration (δ) were 24 ms and 7 ms, respectively. Number of directions scaled with b-values, and the b=0 images were interleaved through the acquisition for improved correction. Data were denoised with MP-PCA[7], [8], [9], [10], corrected for drift, outliers (SOLID[11]), and distortions from susceptibility, motion, and eddy currents were corrected using FSL’s topup[12], [13] and eddy[14]. Gradient non-linearity was corrected using MATLAB, and Gibbs ringing was removed using MRtrix3[15].
For the TADRED, SANDI[16] model parameter maps were generated from the full dataset with a random forest regression as base parameter maps, using the SANDI MATLAB Toolbox[17].
Data Subsampling and Analysis:
Two subsampling approaches were used to optimally subsample the dMRI data: uniform subsampling and TADRED framework[4]. In the first approach, data were reduced uniformly by 10%, 30% and 50% from each b-shell, ensuring that the remaining directions are still distributed uniformly on the unit sphere. For the latter approach, the dMRI data, the generated SANDI maps and acquisitions parameters were concatenated and provided as an input to TADRED framework. The TADRED subsampling and task networks were trained on these SANDI maps using 84% of the data for training and 8% each for validation and testing. Finally, TADRED generated desired subsampled datasets without any constraints from the fully sampled dMRI data. SANDI parameters maps were generated for uniformly and TADRED-subsampled datasets in the same way as the full dataset, which were compared to ‘gold-standard’ maps to evaluate the accuracy and effectiveness of each subsampling method. Fig. 1 shows the subsampled diffusion directions across several b-shells for both uniform and TADRED approaches. Notably, TADRED removed more directions at the intermediate b-shell (b = 2.4 ms/μm²).
Fig. 2 displays the TADRED-predicted SANDI maps alongside ‘ground-truth’ maps for subsampling rates of 10% (A) and 50% (B). Difference maps highlight parameter discrepancies between TADRED outputs and the ground truth.
Fig. 3 compares SANDI model parameter maps derived from the uniform and TADRED-subsampled datasets, illustrating variations between the two approaches.
Fig. 4 presents the mean squared error (MSE) and mean absolute error (MAE) of the SANDI parameters, averaged across all brain volumes. TADRED test results show less than 20% error within brain tissue, with higher errors primarily observed in cerebrospinal fluid regions. In general, TADRED demonstrates lower errors in the difference maps of SANDI parameters across all subsampling rates compared to uniform subsampling (Fig. 3). However, at the 50% subsampling level, errors in fneurite and Rsoma are higher than those from uniform subsampling (Fig. 4); however, the higher error in the voxels originates in the CSF. In this study, we only utilised the TADRED subsampling network to focus on its protocol optimisation capabilities, applying the same model fitting method to both optimisation approaches. Future work will incorporate the TADRED task network to directly estimate the SANDI parameters. TADRED outperformed uniform subsampling in estimating SANDI parameters, showing lower error rates and promising potential for efficient dMRI protocol optimization in future clinical and research applications.
Kadir ŞIMŞEK (Cardiff, United Kingdom), Marco PALOMBO, Muhammed BARAKOVIC, Stefano MAGON, Jens WUERFEL, Derek K. JONES, Paddy SLATOR
11:08 - 11:10
#47583 - PG209 Automated Discovery of Pulsed Saturation Transfer Acquisition Protocols using an Autodifferentiable-Solver Fused with a Quantification Network (AutoPulST).
PG209 Automated Discovery of Pulsed Saturation Transfer Acquisition Protocols using an Autodifferentiable-Solver Fused with a Quantification Network (AutoPulST).
Saturation transfer (ST) MRI has shown promise in various molecular imaging tasks [1–5]. However, its traditional contrast-weighted form is affected by multiple confounding tissue and pulse-sequence parameters [2,6]. Quantitative ST techniques, such as QUESP, yield improved specificity, yet assume steady-state conditions that require a relatively long acquisition [6,7]. ST Magnetic resonance fingerprinting (MRF) overcomes these challenges by matching transient experimental signals to a precomputed dictionary [8–13], predominantly under preclinical settings or continuous wave (CW) acquisition. However, to render ST MRF a clinically viable method, the acquisition protocol must be shortened, optimized for clinical scanners, and accommodate rapid pulsed wave (PW) saturation [12]. Deep learning-based MRF pipelines [14, 15] can efficiently generate optimized acquisition protocols. Yet, to date, they were only compatible with CW acquisition, due to their inherent reliance on analytical solutions of the Bloch-McConnell (BM) equations. Here, we developed a fully differentiable framework for Automatic end-to-end discovery of Pulsed ST MRF sequences (AutoPulST).
A differentiable ST computational graph was implemented in JAX [16], unlocking the ability to rapidly extract the numerical solution of the BM equations. The solver was wrapped within an automatic acquisition protocol discovery framework [14], where a multi-layer perceptron quantification network is jointly trained (Fig. 1). The pipeline was implemented for discovering protocols composed of 4, 8, or 16 raw MRF images (representing a 1.9-7.5 fold acceleration). Before each optimization attempt, a baseline reference protocol was randomly generated.
Imaging was performed at 7T (Terra, Siemens Healthineers) and 3T (Prisma, Siemens Healthineers). Pulsed ST-MRF sequences were automatically generated and implemented for two imaging scenarios: L-arginine phantom imaging (25-100 mM, pH 4–6, chemical shift = 3 ppm, room temperature, 7T) and semi-solid MT brain imaging of healthy volunteers (3T, following IRB approval and informed consent). The backbone pulse sequence used a spin lock saturation train (13x100 ms, 50% duty-cycle), which varied the saturation pulse power and frequency offset. A 3D centric reordered EPI (3T) or GRE (7T) readout module was used [17, 18]. For comparison, a previously established MRF protocol was implemented, which acquires 30 raw images within 143.4 s (3T) or 325.6 s (7T) [19]. In vitro imaging at 7T: the quantitative parameter maps obtained using AutoPulST demonstrated improved SNR compared to the random reference protocols and were visually similar to the gold standard (Fig. 2A). The absolute percent errors obtained by the full AutoPulST pipeline (namely, simultaneous optimization of the acquisition and quantification network) were more than 50% lower than those obtained by the reference random scan (p<0.001. Fig. 2B).
Human brain imaging at 3T: A clear visual improvement was obtained using AutoPulST generated protocols (Fig. 3) compared to the random acquisition protocols. The AutoPulST optimized protocols provided statistically significant quantification improvement (p<0.001 across 12 optimization attempts, Fig. 4) compared to the random alternatives and were acquired within merely 19.1, 38.2, and 76.5 s. In this study, only two acquisition parameters (B1 and Δωrf) were optimized. Nevertheless, we expect that incorporating additional parameters (e.g., saturation pulse time, recovery time, etc.) could further improve the quantification ability. We demonstrate a differentiable, end-to-end optimization framework for rapid pulsed ST-MRF. We expect this approach to play a key part in the translation efforts of quantitative and rapid ST imaging.
Nikita VLADIMIROV (Tel-Aviv, Israel), Edna FURMAN-HARAN, Simon WEINMÜLLER, Moritz ZAISS, Hadar KOLB, Or PERLMAN
11:10 - 11:12
#47789 - PG210 Assessment of Uncertainty and Calibration of Voxel-Wise Supervised Modeling in IVIM.
PG210 Assessment of Uncertainty and Calibration of Voxel-Wise Supervised Modeling in IVIM.
Intravoxel Incoherent Motion (IVIM) is a diffusion-weighted (DW) MRI technique that models signal decay from tissue diffusion and capillary blood flows [1].
Deep Neural Networks (DNNs) have emerged as a powerful alternative to traditional fitting methods, offering fast and robust inference with respect to conventional non-linear least squares (LSQ) and Bayesian approaches [2], [3].
DNNs typically provide only point estimates without uncertainty, which can help in understanding the impact of noise and guiding experimental design [4]. This study, funded by a PRIN 2022 project [5], introduces a set of methods and metrics, based on the use of Mixture Density Networks (MDNs) [6] with Deep Ensembles (DEs) [7] enabling, in a supervised voxel-wise setting, the estimation of both aleatoric (AU) and epistemic uncertainty (EU) and model calibration in IVIM.
We trained our networks using simulated images based on the Shepp-Logan phantom in MATLAB, covering a wide range of IVIM parameters: D (0-0.003 mm²/s), f (0-0.4), and D* (0.003-0.2 mm²/s). The images were computed at the same b-values as those used in our in vivo acquisitions, employing the model equation:
S(b) =S₀[f · e–b·D* + (1 – f) · e–b·D ]
Rician noise was incorporated with SNRs of 25, 50 and 100, for a total number of 6000 phantoms.
We used one in-vivo dataset for testing the networks in a real scenario: mouse brain data (n=6) acquired using a 7T Bruker Biospec using EPI-DWI with 14 b-values (0-1000 s/mm²). Acquisition parameters: TR = 3 s, TE = 61 ms, 2 segments, 2 repetitions, FOV =15 × 15 mm, MTX = 76 × 76, slice thickness = 0.5 mm, δ = 5.8 ms, ∆ = 50 ms.
Architecture
We implemented three voxel-wise models: an MLP, an MDN with either three or one Gaussian. All models used two hidden layers with ELU activations and a number of neurons equal to the number of b-values. Inputs were IVIM signals at each b-value, normalized by the signal at b = 15 (excluding b = 0 due to artifacts in-vivo). Sigmoid activations were applied to the output and scaled to the physiological ranges of the IVIM parameters.
To capture overall uncertainty, we trained an ensemble (M=5) of MDNs with different random initializations. For each MDN in the ensemble, the predictive mean and variance were computed as a weighted average of its Gaussian components. Then, AU was computed as the average predictive variance across ensemble members, while EU was estimated as the variance of the predictive means of each MDN [7].
Experimental
The dataset was split into 70% training, 20% validation, and 10% test sets, maintaining the SNR distribution across subsets. Adam optimizer (learning rate 0.0001, batch size 32) for up to 600 epochs, with early stopping (patience of 35) based on validation loss was used. The MLP used Mean Squared Error loss, while probabilistic models optimized the negative log-likelihood [8], [9].
For simulations, accuracy was assessed using the Median Absolute Error (MdAE), the Relative Median Bias (MdB) and Robust Coefficient of Variation (RCV) [10]. To quantitatively evaluate the quality of uncertainty estimates, we employed calibration reliability diagrams, and the Continuous Ranked Probability Score (CRPS) [11], with lower CRPS indicating better uncertainty estimates. Tab.1 shows the accuracy metrics on the simulation test set across different SNR levels. MLP, MDN, and Gaussian models perform similarly and outperform the Bayesian fitting method.
Fig.1 displays an MDN prediction on a Shepp-Logan phantom at SNR 50, where AU dominates due to noise (mean AU: 4.6×10⁻⁵ mm²/s for D, 0.02 for f and 0.02 for D*), while EU remains low (mean EU: 4.7×10⁻6 mm²/s for D, 0.001 for f and 0.001 for D*).
The reliability diagrams of MDN and Gaussian models are reported in Fig.2, showing better calibration for MDN, as evidenced by a smaller miscalibration area. CRPS values also favor MDN, with slightly lower scores: D (5×10⁻⁵ vs 6×10⁻⁵ mm²/s), f (0.019 vs 0.021), and D* (0.021 vs 0.022 mm²/s).
Fig.3 shows an MDN prediction on an in-vivo slice, where EU - especially for f - is notably higher (around 0.02) than in the simulated case (Fig.2), reflecting the model’s increased uncertainty on real data. We adopt, for the first time in IVIM, probabilistic models like MDNs with DEs to estimate both AU and EU. EU, even though much lower than AU in our study, is often overlooked in quantitative MRI but is crucial for detecting out-of-distribution data. The elevated EPU for f in Fig. 3 may suggest a mismatch between the broad simulation range (0–0.4) and in-vivo values. Quantitative metrics for calibration and CRPS allowed to assess the better reliability of uncertainty estimation of MDN. Our results demonstrate the value of probabilistic modeling for IVIM parameter estimation and uncertainty quantification. Future work will focus on incorporating spatial context via convolutional architectures (e.g., CNNs) and exploring alternative probabilistic approaches such as Normalizing Flows.
Nicola CASALI (Milan, Italy), Alessandro BRUSAFERRI, Giuseppe BASELLI, Stefano FUMAGALLI, Micotti EDOARDO, Gianluigi FORLONI, Riaz HUSSEIN, Giovanna RIZZO, Alfonso MASTROPIETRO
11:12 - 11:14
#47834 - PG211 Radiomics-Based Feature Extraction from DCE-MRI Analysis for Differentiating True Progression and Pseudoprogression in Glioblastoma.
PG211 Radiomics-Based Feature Extraction from DCE-MRI Analysis for Differentiating True Progression and Pseudoprogression in Glioblastoma.
Glioblastoma (GBM), which receives a grade IV classification from the World Health Organization (WHO), is the most prevalent malignant brain tumor in adults [1]. Differentiating between TrueProgression (TP) and Pseudoprogression (PsP) remains a diagnostic challenge, as both appear to be similar on structural MRI sequences like T1 post-contrast (T1pc) and T2 FLAIR. Misclassification of TP or PsP may result in unnecessary interventions or delayed treatment actions, which diminishes the overall patient outcome [2]. Dynamic contrast enhanced MRI (DCE-MRI) based pharmacokinetic (PK) modeling adds important information about tumor vascularity and perfusion. This study employs a parsimonious DCE-MRI model to derive biologically relevant vascular parameters (Ktrans, Ve, and Vp), which are further analyzed through radiomic feature extraction using PyRadiomics to enhance the detection of subtle patterns[3]. Machine learning models were assessed based on their capabilities to classify TP vs. PsP. Lasso, elastic net, and ridge regression were used for feature selection to enhance model performance.
The study featured a private DCE-MRI dataset from the University of Pennsylvania, USA and shared with TCG CREST (data sharing ID: RIS76150), containing 57 GBM cases with 38 TP and 19 PsP. Prior to image processing, a parsimonious modeling approach was used to fit DCE-MRI data and extract pharmacokinetic parameters. Tumors were segmented into enhancing, non-enhancing, and edema regions using the nnU-Net algorithm [4]. The dataset was split into 70% training and 30% testing sets, and SMOTE[5] was applied to correct the 2:1 class imbalance between TP and PsP. Five pharmacokinetic models (Non-linear Tofts, Extended Tofts, Shutter-Speed, 2CXM, and 3S2X) were voxel-wise fitted to generate R² and AIC maps[6].The model with the lowest AIC at each voxel was selected as the best fit. A parsimonious model was then derived by aggregating these voxel-wise best fits to estimate Ktrans (volume transfer constant), Ve (extravascular extracellular volume fraction), and Vp (plasma volume fraction) [7] as shown in Fig 1. Total of 280 radiomic features (140 for enhancing and 140 for non-enhancing regions) were extracted from the pharmacokinetic maps to quantify tumor heterogeneity. Four machine learning classifiers were trained on the selected features: Random Forest (RF), Support Vector Classifier (SVC), Logistic Regression, and XGBoost. Each model were trained and evaluated with and without feature selection to assess the impact of dimensionality reduction on classification performance. The classification performance was assessed using Accuracy and F1 Score. The pharmacokinetic parameters were analyzed separately to determine their predictive power. To improve classification performance, we applied three different feature selection techniques—LASSO, Ridge, and Elastic Net—to identify the most relevant features. Fig. 2 presents the best-performing models for each feature selection method and pharmacokinetic parameter, along with their corresponding accuracy and F1-score. The highest-performing model in each category is highlighted in bold. ROC curve for all the models after feature selection using Elastic Net is shown in Fig 3 for the Ktrans parameter. For each estimated pharmacokinetic parameter, the important features after elastic net and gini importance (Fig 4) included: original shape - least axis length, GLSZM size zone percentage, original shape - sphericity, first order -variance, root mean squared, total energy, range. Random Forest remained a strong performing model, consistently performing well due to its abilities to address underlying non-linearity and feature interactions. The ensemble
approach of Random Forest also assisted in minimizing overfitting while working with a small dataset. In a similar manner, SVC (with RBF kernel) produced strong results,
especially for Ve, likely due to the manner in which it can build non-linear decision boundaries in high dimensional feature spaces. Elastic Net addressed the challenges of overfitting with the balance of L1 and L2 regularization while selecting the most informative features. This study supports that the maximum classification performance occurred when Elastic Net, with Random Forest, was applied to Ktrans, achieving F1-score of 92.86% with accuracy of 88.89%. The present study helps advance the use of machine learning applications in medical imaging by considering pharmacokinetic parameters during classification of the tumor type. An important limitation of this study is the small sample size which limited appropriate validation and increased the risk of overfitting. Future studies can explore multiparametric approach combining multiple features may further improve the robustness of tumor classification models. Additionally, as dataset sizes increase, deep learning architectures can increase the classification accuracy.
Sourav BASAK (Kolkata, India), Akashleena CHATTERJEE, Subhanon BERA, Gabriela W KOSTRZANOWSKA, Archith RAJAN, Harish POPTANI, Sanjeev CHAWLA, Sourav BHADURI
11:14 - 11:16
#47904 - PG212 Improving Cerebral Perfusion Estimation in ASL Using Physics-Informed Neural Networks: A Simulation Study.
PG212 Improving Cerebral Perfusion Estimation in ASL Using Physics-Informed Neural Networks: A Simulation Study.
Arterial Spin Labeling (ASL) is a non-invasive MRI method for quantifying cerebral perfusion via magnetically labeled blood water. Time-encoded pseudo-continuous ASL (te-pCASL) with Hadamard encoding enhances signal efficiency and enables simultaneous cerebral blood flow (CBF) and arterial transit time (ATT) estimation [1]. The Buxton model describes the ASL signal evolution and is widely used for perfusion quantification [2], though nonlinear least squares (NLLS) methods often yield unstable parameter maps under low signal-to-noise conditions due to sensitivity to noise and initialization.
Physics-Informed Neural Networks (PINNs) offer a robust alternative by embedding governing physical models into the learning process, improving resilience to noise and data sparsity [3]. Their applications in biomedical imaging are expanding, including cardiovascular modeling [4], perfusion CT [5], and neonatal ASL [6]. Building on [6], we extend the PINN framework to te-pCASL and assess its performance in a synthetic simulation study.
We designed a two-stage PINN framework trained on synthetic te-pCASL data generated using spatially heterogeneous CBF, ATT, and equilibrium magnetization (M₀) derived from Boston ASL Template and Simulator [7]. A central slice was extracted and resampled to match the voxel size of real ASL datasets (3x3x7 mm3). The actual signals were simulated using a custom implementation of the Buxton model, accounting for sub-bolus-specific labeling durations (Figure 1). Gaussian noise was added to yield low temporal signal-to-noise ratio conditions (tSNR = 1.5 and 0.5), following methods from prior studies [8].
The model comprised two coupled SIREN-based Multi-Layer Perceptrons (MLPs): a data-fitting network (7 layers, 256 units) mapping spatial coordinates (x, y) and time to ASL signal, and a physics-based network (4 layers, 128 units) mapping (x, y) to CBF and ATT values (Figure 2).
To consider the dependence of bolus duration on acquisition time in absence of an analytic expression, a fifth-order spline was used to model this relationship, and its derivative was included in the Buxton model derivative during training to better reflect time-encoding dynamics.
Training minimized a hybrid loss combining mean squared error (MSE) between predicted and simulated signals, and MSE between time derivatives of the data-fitting network and those obtained from the Buxton model derivative using predicted parameters. Optimization used Adam (learning rate=0.0001) for 30,000 epochs with a batch size of 150. At inference, only the physics-based network was used for parameter estimation.
As a baseline, we used a regularized NLLS estimator with soft-L1 loss, physiological constraints, and optimization via Dogbox. It minimized residuals between signals and Buxton model predictions, with mild regularization on ATT deviations from initialization.
Performance was evaluated using Structural Similarity Index Measure (SSIM), Pearson Correlation Coefficient (r), and Percentage Relative Error (PRE), along with visual inspection for qualitative comparison. At tSNR = 1.5, both methods performed similarly for CBF. However, PINN significantly outperformed NLLS in ATT estimation, a parameter more sensitive to noise (Table 1). At tSNR = 0.5, PINN’s advantage was even more pronounced, achieving higher SSIM and r values for both parameters.
Qualitatively, PINN produced smooth, physiologically explainable maps (Figure 3). While ATT overestimation persisted, PINN better preserved spatial structure. Incorporating the spline correction reduced ATT bias, indicating better modeling of bolus-specific dynamics, though a residual bias remained. NLLS maps, in contrast, showed higher noise and voxel-wise variability, particularly for ATT. Embedding the Buxton model in a PINN framework enhances robustness in perfusion quantification under noise. In particular, ATT estimation benefits significantly, addressing limitations of conventional methods. The spline-based correction partly mitigated ATT bias, supporting the hypothesis that bolus duration variability influences estimation accuracy.
Nevertheless, the remaining bias suggests additional sources of mismatch, possibly due to simplifications in the kinetic model or training dynamics. Future work will explore further physical model refinements and apply the framework to real ASL datasets. In conclusion, we propose a PINN-based framework for ASL quantification, integrating the Buxton model as a physical constraint. Compared to NLLS, our method improves ATT estimation and noise robustness, while maintaining physiologically plausible spatial patterns. This work lays the foundation for extension to 3D and clinical ASL studies.
Alessandro GIUPPONI (Padova, Italy), Chiara DA VILLA, Mattia VERONESE, Marco CASTELLARO
11:16 - 11:18
#46950 - PG213 Uncovering Alzheimer’s Disease Prediction Strategies of Convolutional Neural Network Classifiers using T1-weighted MRIs and Spectral Clustering.
PG213 Uncovering Alzheimer’s Disease Prediction Strategies of Convolutional Neural Network Classifiers using T1-weighted MRIs and Spectral Clustering.
Convolutional neural networks (CNNs) have shown strong performance in classifying Alzheimer’s disease (AD) from T1-weighted (T1w) MRI [1], yet their decision-making remains largely opaque [2]. Ensuring decisions are not driven by spurious features is critical for clinical applications.
Prior work [3] indicated that CNNs favor skull-stripped over full images for T1-based AD classification. However, how learned features are spatially organized under different preprocessing pipelines remains unstudied. Spectral clustering (SC) [4] can group similar heatmaps and reveal dominant decision patterns beyond individual cases.
This study applies Spectral Relevance Analysis (SpRAy) [5] to Layer-wise Relevance Propagation (LRP) [6] heatmaps in a CNN trained on T1w MRI. We assess whether relevance patterns vary across preprocessing methods and identify brain regions that consistently drive classification, allowing insights into model behavior and potential preprocessing biases.
Participants.
From the ADNI database (https://adni.loni.usc.edu), we selected 1,042 AD and 2,227 normal control (NC) T1w MRIs acquired sagittally at 3T with consistent resolution. A balanced dataset was constructed with 990 images each from 159 AD and 201 NC participants (propensity-score matched by age and sex) [7]. Scans were split into training (70%), validation (15%), and test (15%) sets with subject-level separation.
Preprocessing.
Images were reoriented, bias-corrected [8], non-linearly registered to MNI space [9], and normalized to the white matter histogram peak. Skull-stripping was performed using FSL-SIENAX [10]. Binarization thresholds of 13.75%, 27.5%, and 41.25% of white matter peak intensity were applied to generate texture-masked images, resulting in eight preprocessing conditions (aligned/skull-stripped × 4 thresholds, see Fig. 1).
CNN Architecture.
We adapted a 3D CNN from [1] with reduced complexity to avoid overfitting. For a schematic overview see Fig. 2.
Training.
Each configuration was trained for 30 epochs (batch size 20) using Adam [11]. Ten random data samplings per configuration were used [12], with three models trained per sample (total: 30 per setup) [13]. The best validation-performing model per setup was used for further analysis.
Heatmap Generation and Spectral Clustering.
LRP (α=1.0, β=0.0) [6] generated voxel-wise relevance maps. Mean heatmaps were computed and thresholded to the top 40% of relevance for inspection.
SpRAy involved:
1. Generating LRP heatmaps,
2. Downsampling to 2 mm resolution,
3. Spectral clustering of relevance maps,
4. Identifying meaningful eigenvalue gaps,
5. Visualizing clusters via t-distributed stochastic neighbor embedding (t-SNE) [14]. Skull-stripped data without binarization (A2) achieved the highest accuracy (81.6%). Other configurations, including 27.5% binarized skull-stripped images (C2) and 41.25% binarized aligned images (D1), performed comparably. For full statistics, refer to [3].
t-SNE visualization (Fig. 3) showed that only model D1 produced heatmap clusters aligning with subject groups (AD vs. NC). This suggests that omitting skull-stripping and using 41.25%-threshold binarization may lead to more positionally distinctive relevance distributions.
Clustered mean heatmaps (Fig. 4) showed D1’s Group 2 highlighting the left insular cortex -a known AD-affected area- while Group 3 included skull regions, pointing to potential reliance on non-brain features. Models A2 and C2 showed less separation and less anatomically interpretable relevance. This study extends prior findings on CNN reliance on volumetric cues [3] by showing how preprocessing impacts relevance patterns. SC enabled group-level analysis of LRP heatmaps, identifying systematic rather than instance-specific decision strategies.
Only D1, the model trained on non-skull-stripped and 41.25% binarized images, revealed clustering aligned with AD vs. NC labels. Its mean heatmaps featured clearer anatomical relevance (e.g., left insular cortex), but also highlighted skull regions, suggesting possible biases. This reveals a trade-off: less aggressive preprocessing may preserve informative structure but also introduce non-brain artifacts.
Importantly, similar classification performance across all configurations obscures these interpretability differences, underscoring the need for explainability techniques in model evaluation [15]. Preprocessing choices, even minor ones, can substantially alter feature attribution. We applied SC to LRP heatmaps from CNNs trained on differently preprocessed T1w MRIs for AD classification. Despite similar accuracies, relevance patterns varied markedly, with the 41.25% binarized model without skull-stripping (D1) showing clearer separation between AD and NC. Our findings stress that preprocessing has a strong impact on interpretability, not just performance. Future work should explore the effects of preprocessing and the integration of quantitative MRI features to improve model transparency and clinical utility.
Christian TINAUER (Graz, Austria), Maximilian SACKL, Stefan ROPELE, Christian LANGKAMMER
11:18 - 11:20
#46479 - PG214 Preliminary Evaluation of Deep-learning Image Reconstruction in Clinical Setting at 7T.
PG214 Preliminary Evaluation of Deep-learning Image Reconstruction in Clinical Setting at 7T.
While deep learning (DL) methods in MR image reconstruction are becoming state-of-the-art, the possibility of DL models altering the appearance of pathology still raises concerns, especially when trained solely with healthy subjects and/or when employed on patients’ datasets acquired with higher undersampling factors [1]. This issue is particularly relevant and not sufficiently examined in the case of clinical acquisitions of the brain with submillimeter resolutions, where undersampling is highly desirable to reduce acquisition time.
In this study, a DL-based reconstruction was investigated for the acceleration of patients’ clinical scans acquired with 0.6 mm isotropic resolution at 7T. Images were retrospectively undersampled to test different accelerations, and image quality and pathology conspicuity were compared against conventional compressed sensing (CS) reconstruction.
Study Population and MR Acquisition
Six patients were enrolled in this preliminary analysis: three patients with different brain tumors and three patients with suspected multiple sclerosis (MS). All patients were scanned at 7T (MAGNETOM Terra, Siemens Healthineers, Forchheim, Germany) using an 1Tx/32Rx RF head coil (Nova Medical, Wilmington, USA). A 3D MP2RAGE research application sequence (resolution = 0.6×0.6×0.6 mm3, matrix size = 384x256x384, TR/TI1/TI2 = 6000/800/2700 ms, TE/echo-spacing = 2.06/6.2 ms, acquisition time = 7:40 min) with a 4x accelerated spiral phyllotaxis sampling was acquired [2–4]. The work was approved by the local ethics committee (Kantonale Ethikkommission für die Forschung Bern, Switzerland, 2020–02902, 2022–00720, 2024-00788). Prior to each examination, written informed consent was obtained.
Image Reconstruction
MP2RAGE acquisitions were also retrospectively undersampled to simulate acceleration factors of R=8 (3:50 min) and R=10 (3:04 min). All images were reconstructed using both a conventional CS reconstruction [5,6] and a 3D DL-based method [7]. The DL technique reconstructs images from undersampled k-space data and coil sensitivity maps using six iterations of data consistency updates and neural network evaluation. The network was initially trained with 5000 pairs from fully-sampled 3D datasets of healthy subjects scanned at 1.5 and 3T, followed by fine-tuning in a self-supervised manner using 1000 pairs of undersampled 3D datasets collected at 7T [8,9] (all MAGNETOM scanners, Siemens Healthineers, Forchheim, Germany).
Image Analysis
A visual inspection and a quantitative quality assessment of retrospectively reconstructed images was conducted by computing the structural similarity index measure (SSIM) and the peak signal-to-noise ratio (PSNR) considering the current clinical protocol (R=4, CS reconstruction) as reference. Reconstructed images for patients with suspected dysembryoplastic neuroepithelial and epidermoid tumor are shown in Figure 1-2. Overall, DL reconstructions exhibit higher SNR than CS reconstructions while preserving tissue contrast. For the highest tested acceleration (R=10), better (Figure 1) or similar (Figure 2) conspicuity of tumor boundaries and higher SNR was observed in the DL reconstruction compared to CS.
Images from two of the MS patients are reported in Figure 3-4. Lesions identified with acceleration R=4 could be visualized at higher acceleration rates. Lesion conspicuity was found to be similar or better in DL reconstructions in comparison to CS.
None of the images reconstructed with the DL reconstruction exhibit hallucinations or misleading artifacts.
For R=8, CS reconstruction achieved an SSIM of 0.99±0.01 and a PSNR of 37.3±0.3 dB, while DL reconstruction yielded an SSIM of 0.99±0.01 and a PSNR of 38.1±0.5 dB. At R=10, CS reached an SSIM of 0.98±0.01 and a PSNR of 35.6±0.2 dB, whereas DL reconstruction achieved an SSIM of 0.99±0.01 and a PSNR of 37±0.4 dB. In this preliminary study, we evaluated the performance of a DL-based reconstruction method trained solely on healthy subjects to reconstruct images of patients with different brain abnormalities. No hallucinations or misleading artifacts resulting from the DL reconstructions were observed. Results were compared to a CS reconstruction, showing that the DL reconstruction exhibits higher SNR, PSNR and equivalent or improved conspicuity in the observed pathologies. These findings suggest robustness of the DL approach across different pathological presentations.
The work conducted in this study is planned to be extended to a larger cohort of patients including radiologist rating of the reconstructed images, while being blind to the acceleration factor or the reconstruction technique, to establish the clinical value of DL reconstruction more comprehensively for 7T neuroimaging. This preliminary study demonstrates the quality and reliability of DL-based MRI reconstruction in the presence of pathology, even at high acceleration rates. In the future, we will validate these results in a larger cohort of patients.
Jocelyn PHILIPPE (Lausanne, Switzerland), Gian Franco PIREDDA, Natalia PATO MONTEMAYOR, Dominik NICKEL, Patrick LIEBIG, Arsany HAKIM, Robin HEIDEMANN, Jean-Philippe THIRAN, Tom HILBERT, Gabriele BONANNO, Piotr RADOJEWSKI, Thomas YU
11:20 - 11:22
#46748 - PG215 Explainable Machine Learning Models for Radiomic-Based Assessment of Glioma Severity Using Multiparametric MRI.
PG215 Explainable Machine Learning Models for Radiomic-Based Assessment of Glioma Severity Using Multiparametric MRI.
Gliomas are primary brain tumors characterized by marked biological heterogeneity, affecting prognosis and treatment strategies [1,2]. Traditional histopathological grading, though informative, is invasive and limited by sampling bias [3,4]. Non-invasive radiomic analysis using MRI has shown promise in assessing tumor grade [5,6]. However, most machine learning (ML) studies frame glioma grading as a classification task, potentially overlooking subtle intra-class variability [7]. In this study, we propose a regression-based ML approach to predict glioma grade as a continuous variable using radiomic features extracted from T1-weighted and diffusion tensor imaging (DTI) to enhance diagnostic precision and interpretability [8,9].
We analyzed MRI data from 36 glioma patients (58.3% low-grade gliomas [LGG], 41.7% high-grade gliomas [HGG]) acquired on a 1.5T scanner. Radiomic features were extracted from normalized T1-weighted images and diffusion maps, including axial diffusivity (AD), radial diffusivity (RD), mean diffusivity (MD), and fractional anisotropy (FA) [10,11]. Feature extraction included intensity, shape, and multiple texture matrices (GLCM, GLRLM, GLSZM, NGTDM, GLDM) using PyRadiomics [12]. Dimensionality reduction was performed using Sequential Feature Selection (SFS) with a Random Forest regressor [13]. A total of 15 ML regression models, including XGBoost, CatBoost, SVM, and neural networks, were trained and optimized via stratified 5-fold cross-validation. Model performance was evaluated using MSE, MAE, and R² scores. SHAP analysis was applied to interpret the contribution of individual features [14], as showed in Figure 1. Among all models, XGBoost achieved the best performance with an MSE of 0.0346 ± 0.0137, MAE of 0.1168 ± 0.0256, and R² of 0.5140 ± 0.3579 (Figure 2). Feature selection identified six key predictors, with texture features from T1-weighted images (e.g., GLRLM short run low gray level emphasis, GLCM contrast) being the most influential (Figure 3). Diffusion-derived features, particularly robust mean absolute deviation from AD maps, added complementary information. SHAP analysis highlighted how selected features differentially contributed to grade predictions across LGG and HGG samples, with interpretable patterns aligning with tumor microstructural complexity (Figure 4) [15,16]. Modeling glioma grade as a continuous variable allowed for finer granularity in characterizing tumor severity, capturing nuances missed by binary classification [17]. Texture features reflecting tumor heterogeneity and shape irregularity emerged as strong predictors. Diffusion metrics contributed additional microstructural information, reinforcing the importance of multimodal integration [18,19]. The regression approach provided accurate predictions and clinical insights, particularly when supported by explainability techniques like SHAP. Differences in hemispheric tumor location between LGG and HGG further support the biological underpinnings captured by the radiomic features [20]. Radiomic features from T1-weighted and DTI sequences, analyzed through ML regression models, enable accurate, interpretable, and non-invasive glioma grading. The XGBoost regressor demonstrated superior performance and SHAP-based analysis enhanced transparency. This continuous-scale grading strategy offers a refined perspective on tumor biology and supports its use in clinical decision-making. Future studies with larger and multi-site cohorts are encouraged to validate the model’s generalizability and explore its integration into routine radiological workflows.
All radiomic processing scripts, ML models, and anonymized datasets used in this study are available from the corresponding author upon reasonable request. Feature extraction was conducted using PyRadiomics v3.0 in Python 3.8, and model development used open-source libraries, including Scikit-learn and XGBoost.
Pamela FRANCO, Cristian MONTALBA (Santiago, Chile), Raúl CAULIER-CISTERNA, Ignacio ESPINOZA, Carlos BENNET, Francisco TORRES, Steren CHABERT, Rodrigo SALAS
11:22 - 11:24
#46455 - PG216 Understanding predictive uncertainty of AI and expert confidence in multiple sclerosis cortical lesion segmentation on MP2RAGE.
PG216 Understanding predictive uncertainty of AI and expert confidence in multiple sclerosis cortical lesion segmentation on MP2RAGE.
Cortical lesion (CL) detection in magnetic resonance imaging (MRI) is a key biomarker for differential diagnosis and disability assessment in multiple sclerosis (MS) [1,2] (Figure 1). Recent deep learning (DL) methods have improved the standardization and accuracy of CL segmentation [3,4]. Trustworthy AI, particularly with uncertainty quantification (UQ), enhances model reliability and robustness, which are critical for clinical use [5]. While UQ is often used to estimate prediction errors, its clinical interpretation remains underexplored. Our prior studies [6,7] linked high lesion-level uncertainty with interpretable imaging features. This study examines how lesion-scale uncertainty relates to expert perception, including lesion type, diagnostic confidence, and segmentation quality.
The expert perception was assessed with the participation of an experienced neurologist (AC, 6 years of MRI experience). Figure 2 outlines the study workflow. We used our previous 3D nnU-Net model [7,8] trained on a clinical MP2RAGE dataset [9,10] with manual CL annotations by consensus between two experts (a medical doctor and the same neurologist; both had 5 years of MRI experience). The dataset included 163 MS patients (train:val:test = 109:13:41; CLs = 857:69:301). Test set lesion uncertainty was estimated using deep ensembles [11], and lesion structural uncertainty (LSU) [12] was computed (range: 0–1; higher - more uncertain). Lesions were grouped into five equidistant LSU bins; up to 23 per bin were randomly sampled, yielding 100 regions of interest (ROIs; Figure 2, Step 2). Due to the skewed LSU distribution, 68 ROIs had LSU > 0.19 (above 75th percentile); 34 of 100 were false positives. The neurologist reviewed each ROI overlaid on MP2RAGE using their preferred viewer, with detailed written instructions. For each ROI, they reported lesion type, confidence (5-point Likert), reasons for reduced confidence (from 13 predefined categories [7]), and segmentation quality (high/moderate/low) using a multiple-choice table-like form (Figure 2, Step 3). We analyzed categorical expert ratings and continuous variables not visible to the neurologist: LSU and lesion segmentation quality, measured by IoUadj between ROI and ground truth masks [13]. Distributions were visualized via violin plots, and group differences were tested using the Mann–Whitney U-test (n > 10). Inverse confidence scores weighted confidence-reducing factors to highlight dominant sources of uncertainty. The average self-reported assessment time was 30 sec/ROI. Most ROIs were identified as leukocortical (n=36) or juxtacortical (n=50), with no significant LSU difference between groups (Figure 3-i). Notably, 9 of 34 false positives were reclassified as true lesions. Among the 66 lesions overlapping with the ground truth, 35 were labeled as non-CL (33 juxtacortical MS, 2 vascular MS lesions). Annotator confidence was most often "slightly" (n=42) or "moderately" confident (n=34) (Figures 3-ii, 4-ii). LSU has significantly different distributions across the perceived segmentation quality (Figure 3-iii) with p < 0.01. There was no statistical difference between evaluated segmentation quality (IoUadj) distributions for “high” vs “moderate” perceived quality (p=0.11), but a clear difference between “moderate” vs “low” (p<0.01). By far, the most influential factor lowering confidence was "unclear cortical involvement" (Figure 4-i). We sampled lesions uniformly across LSU values to explore a possible one-to-one mapping with annotator confidence levels. However, the estimated annotator confidence distribution is also skewed, suggesting that this direct mapping is not viable. On the other hand, most of the sampled ROI had high LSU (above the 75th percentile) and were also deemed difficult by the neurologist, who was mostly “moderately confident” and changed his opinion about 44 of 100 ROIs. "Unclear cortical involvement" emerged as the primary reason for diagnostic uncertainty, consistent with the known challenges of CL detection in 3T MRI [14] and our prior uncertainty-focused work [7]. The observed association between LSU and perceived segmentation quality corroborates previous findings [12] and supports its utility in clinical settings. This study advances the understanding of how predictive uncertainty aligns with expert confidence in MS cortical lesion segmentation. Our results demonstrate that lesion structural uncertainty from deep ensembles is a promising tool for identifying clinically ambiguous lesions and guiding expert review without ground truth, showing a potential for clinical decision support and targeted review. Future work should expand expert participation to validate these findings and enhance generalizability.
Nataliia MOLCHANOVA, Alessandro CAGOL, Delphine RIBES, Pedro M. GORDALIZA, Mario OCAMPO– PINEDA, Matthias WEIGEL, Xinjie CHEN, Adrien DEPEURSINGE, Granziera CRISTINA, Müller HENNING, Meritxell BACH CUADRA (Lausanne, Switzerland)
11:24 - 11:26
#47361 - PG217 Deep editable network for automatic and interactive segmentation of skeletal muscle MRI.
PG217 Deep editable network for automatic and interactive segmentation of skeletal muscle MRI.
Quantitative MRI provides increasingly relevant biomarkers for the characterization and monitoring of the musculoskeletal system. Such quantitative image analyses generally require delineating regions of interest (ROI) to separate the different muscles or muscle groups.
Lately, AI models achieved excellent segmentation quality in a small fraction of the time taken by manual segmentation. Despite their efficiency, these automatic methods often generate partially erroneous segmentations, especially in the presence of important pathological involvements such as fatty replacements. In this case, the only way to correct errors is by manual segmentation, which can be very time-consuming depending on the size of the image and the number of errors.
Based on the work of Diaz Pinto et al.[1], we propose a method for semi-automatic 3d muscle segmentation where users can correct errors by providing guidance to the network in the form of clicks or scribbles. On a database of MRI acquisitions or healthy and pathological legs, we trained and tested a modified nnU-net[2] model, including automatically generated clicks within the training loop, and compared its performance to that of the unmodified fully automatic nnU-net model. Initial results show that the proposed model provides a fully automatic segmentation comparable to the non-interactive nnU-net model and, in addition, allows efficient improvements through simple user interactions.
For this study, both training and test sets were composed of 20 MRI acquisitions of legs of consenting volunteers and neuromuscular disease patients, acquired at 3T (PrismaFit, Siemens Healthineers). Images were 3d Dixon acquisitions of 64 slices of 224 x 224 pixels, with TE = 3.95 ms. In both datasets, we selected equal numbers of cases with low and high degrees of fatty replacements. Ground truths were manually delineated for 9 labels : background, tibialis anterior (TA), extensor digitorum (ED), peroneus (PER), deep layer of posterior compartment (DLPC), popliteus (POP), soleus (SOL), lateral gastrocnemius (GAS_LAT), medial gastrocnemius (GAS_MED), by medical experts in 1 out of 4 slices.
Using the nnU-Net framework, we modified the training pipeline to introduce simulated clicks using a method similar to that of Sakinis et al.[3]. In each training batch, clicks are progressively and randomly added into dedicated input channels with probabilities depending on the size of the mislabeled regions. Following Diaz-Pinto et al.[1], simulated clicks are added in only 80 % of the batches to maintain performance without click. We also used a modified loss (Figure 1) that forces the network to focus at first on the click-less segmentation and to prioritize click influence in later epochs.
We trained our model on a Nvidia RTX A6000 using 5-folds cross validation, with each fold running for 800 epochs each. An example of the incremental correction with manual clicks of a test set image is given in Figure 2.
For the testing part, we simulated 20 successive clicks based on the ground truth, computing relevant metrics for each prediction for all labels. The measured metrics were: the Dice similarity coefficient (mean, per label mean, mean of worst label), the maximum error distance (the distance to the border of the innermost pixel of the mislabeling error mask) and the click influence (the number of updated pixels). We also trained and tested an unmodified nnU-Net on the same dataset as benchmark (the “no-click” model). Comparison between models were assessed with paired t-tests. Figures 3 and 4 show that the 0-click and no-click models have equivalent performance in terms of maximum error distance, Dice of worst label, mean Dice, and per-label Dice. As the number of clicks increase, the mean Dice and maximum error distance become significantly better than for the no-click model.
The label frequency plot shows that the larger regions (background, DLPC, SOL) tend to be prioritized for the clicks, while smaller muscles are less often selected. The click influence plot indicates that initial clicks tend to have more influence than later clicks, the latter converging towards about 500 pixels. These results show that adding corrective guidance to the nnU-net model does not penalize the fully automatic segmentation, while automatically added clicks significantly improve the segmentation quality.
We used the nnU-Net framework for practical reasons, but this method can be used with any neural network architecture. Alternative architectures such as transformers could potentially improve segmentation performance.
The above results were based on simulated clicks. Using actual user-guidance provided by medical experts is the logical next step to prove the relevance of the proposed method. After further validation, this approach could be implemented into a simple user-interface to lighten the workload in research and clinical settings, making it possible to generate and correct a full 3D image segmentation in a few minutes.
Louis RIGLER (Paris), Jean-Marc BOISSERIE, Sophie JOUAN, Pierre-Yves BAUDIN
11:26 - 11:28
#47303 - PG218 Uncertainty in Deep Learning of DCE-MRI Parameter Estimation.
PG218 Uncertainty in Deep Learning of DCE-MRI Parameter Estimation.
Dynamic contrast-enhanced (DCE) MRI helps detect and characterize diseases, like cancer and neurodegenerative disorders, by quantifying tissue perfusion. However, conventional methods often yield noisy parameter estimates. Recently, deep learning has emerged as an alternative, offering more accurate and precise parameter estimates. However, even in regions of uncertainty, these deep-learnt parameter-maps are visually appealing , leading clinicians into a false sense of security.
Incorporating uncertainty estimates in a deep learning framework can inform clinicians about trustworthiness. Moreover, understanding whether uncertainties stem from noise in the data (aleatoric) or model limitations (epistemic) can inform us whether we need better data or models. For example, in case of out-of-distribution (OOD) effects, epistemic uncertainty may be reduced by offering more varied training examples.
Therefore, we propose an uncertainty-aware neural network, hypothesising it can detect aleatoric uncertainties (H1) and that a deep ensemble can capture epistemic uncertainties (H2). Finally, we demonstrate how this model’s uncertainties can be visualized in vivo.
We used the extended Tofts model to simulate 100,000 pharmacokinetic concentration-time curves with varying noise levels. This synthetic dataset was split into training, validation, and test subsets.
To test H1, we extended a previously developed network (DCE-NET) by incorporating mean-variance estimation (MVE), enabling the model to output both pharmacokinetic parameters and associated uncertainty. We evaluated whether predicted uncertainties correlated with actual errors using quartile-based stratification and statistical comparisons of error distributions.
To test H2, we trained an ensemble of ten independently initialized MVE-DCE-NET models. Each network’s output was treated as a sample from a posterior distribution, allowing us to compute both predictive mean and epistemic uncertainty. To simulate OOD effects, we intentionally excluded certain parameter ranges from the training and validation data while keeping them in the test set. This allowed us to observe whether the ensemble detected unfamiliar parameter regimes through increased uncertainty.
Finally, we developed a visualization method for uncertainty-aware parameter maps. These maps display predicted parameter values and visually encode confidence by superimposing controlled noise that scales with predicted uncertainty. As a proof of concept, we applied our method to in vivo DCE-MRI data from two healthy volunteers. The uncertainty-aware networks showed more accurate pharmacokinetic parameter predictions than the conventional network (Figure 1A). Larger aleatoric uncertainties predicted by MVE-DCE-NET correlated with a larger spread in parameter errors (Figure 1B). Paired Levene’s test indicated significant differences in error variances across uncertainty quartiles, confirming H1. Additionally, a positive correlation between median predicted uncertainty and error further supported this hypothesis (Figure 2).
Evaluation of epistemic uncertainties revealed that the ensemble effectively identified unseen parameters in the test dataset (Figure 3), with lowest uncertainties for well-represented values and highest for unrepresented ones. Uncertainty was also high for low ke and ve. As curves with little tissue uptake can be explained both with a low ke (and arbitrary ve) or low ve (and arbitrary ke), there is a large uncertainty on ke and ve in those situations.
With no noise, in vivo parameter maps look smooth and trustworthy (Figure 4). However, as the noise scaling parameter increased, the uncertainty-informed maps displayed more noise in uncertain regions, making it clear to the viewer which regions are less trustworthy. This study presents MVE-DCE-NET, an uncertainty-aware neural network designed to predict pharmacokinetic parameters along with their aleatoric and epistemic uncertainties. Our findings demonstrate that MVE-DCE-NET effectively highlights high aleatoric uncertainties when estimation errors are large and shows low uncertainty when errors are low, making results interpretable. Additionally, deep ensembles identified underrepresented data with high uncertainties, which could support create balanced datasets with equal representations of healthy and diseased tissues. If certain tissues exhibit parameters not present in the training data, our approach can flag these tissues, encouraging further examination and reducing reliance on uncertain predictions. This work introduces MVE-DCE-NET, an uncertainty-aware model that enhanced DCE-MRI parameter reliability, distinguishing between inherent data noise and model limitations. This approach can support clinicians in making informed decisions by highlightening prediction confidence and potential outliers.
Natalia KOROBOVA (Amsterdam, The Netherlands), Jonas VAN ELBURG, Mohammad ISLAM, Marian TROELSTRA, Oliver GURNEY-CHAMPION
11:28 - 11:30
#47669 - PG219 Deep Learning based acceleration of Prostate DWI on a 1.5T MR-Linac and assessment of ADC bias and repeatability.
PG219 Deep Learning based acceleration of Prostate DWI on a 1.5T MR-Linac and assessment of ADC bias and repeatability.
Magnetic resonance-guided radiotherapy (MRgRT) enables superior soft-tissue visualisation and daily adaptive treatment planning. Online adaptation can extend treatment sessions by 20–30 minutes, with high-resolution imaging potentially further contributing to this delay [1, 2]. Apparent diffusion coefficient (ADC) maps derived from diffusion-weighted imaging (DWI) show promise as a reliable biomarker for dose escalation based on tumour response [3]. Strategies to accelerate acquisition while preserving image quality are key for improving workflow efficiency for MRgRT where imaging enables online treatment adaptation, such as identifying targets for dose escalation. Although current workflows may accommodate longer scans, future optimizations in radiotherapy delivery will require that imaging speed also keep pace. This study aims to accelerate diffusion imaging for MRgRT using deep learning to generate high-quality DWI and assess the bias and repeatability of the resulting quantitative ADC maps.
This study analysed DWI from 11 prostate cancer patients enrolled in the HERMES clinical trial (REC 20/LO/1162) [4], treated on a 1.5T Unity MR-Linac (Elekta AB, Stockholm, Sweden). The cohort included patients from both the two-fraction (2#) (P1 – P6) and five-fraction (5#) (P7 – P11) arms, with a median age of 72 years (range: 53–77). All patients received neoadjuvant and concurrent androgen deprivation therapy. DW images were acquired using single-shot echo planar imaging with b-values(averages) of 0(6), 30(6), 150(6), and 500(14) s/mm² based on an MR-Linac consortium recommended consensus protocol [5].
A deep learning model [6] based on a U-Net architecture was trained to produce high-quality DWI from noisy input data [Fig. 1]. Training was performed over 70 epochs using the Adam optimizer (learning rate 1e−4), with mean absolute error (MAE) as the loss function and a batch size of 20.
Each training scan consisted of 15 transverse slices, with 30 training samples per slice assembled from different diffusion averages, resulting in 450 training samples per scan. Each sample was represented as a [224 × 224 × 12] volume, where the 12 input channels comprised the current slice, preceding and following slices, and images from the four b-values. For the models, the input at each b-value was a directionally averaged trace image. The specific acquisition or means of any two or three acquisitions was/were randomly selected per sample. In total, 25 scans (11,250 samples) were used for training and 5 scans (2,250 samples) for validation.
A five-fold cross-validation was used for the best model. For Fold 1, patient P1 was used for testing, P6 and P10 for validation, and the remaining patients (P2–P5, P7–P9, and P11) for training. For the subsequent training folds, the other 2# patient data were rotated to be test data.
Three model configurations were evaluated, differing in diffusion input averaging: all-direction input using single, two and three averages respectively:
1. AD_avg1
2. AD_avg2
3. AD_avg3
Quantitative evaluation included voxel-wise fit of ADC of whole prostate (WP) and comparison between model outputs and ground truth (GT). The ADC repeatability coefficients (RC) of WP and gross-tumour volume (GTV) were calculated. Fig. 2 shows a representative transverse slice for visual comparison of GT, inputs and the predicted images.
MAE and RMSE between predicted and GT ADC are reported in Table 1. AD_avg3 had the lowest error, followed by AD_avg2.
Box plots of WP ADC distributions showed good agreement between model predictions and GT, with the models showing a narrower distribution [Fig. 3]. AD_avg2 had a negative bias for the tested patient.
RC Calculation: Absolute ADC RCs in 10-6 mm2/s (relative RC) for the full acquisition were 483 (28.0%), and 234 (12.8%) for GTV and whole prostate respectively. In the predicted ADC maps of AD_avg1 model, these were improved to 181.83 (10.4%) and 180.48 (9.5%). We demonstrate that deep learning can generate high-quality DWI from subsampled data on the MR-Linac. Specifically, model trained with only one average from all-direction data (AD_avg1) produced results visually comparable to full averaging, reducing acquisition time to 36 seconds from the current 4:12 minutes. This model achieved superior ADC repeatability compared to the full acquisition ground truth for this dataset. There is a clear trade-off between denoising performance and acquisition time. While AD_avg3 achieves the lowest error, AD_avg1 offers substantial scan time reduction with only a marginal increase in error, suggesting it may be preferable in a time-constrained clinical setting. Deep-learning accelerated DWI makes feasible shorter scan times, improved patient comfort, and better workflow compatibility for diffusion imaging in MRgRT without compromising image quality.
Prashant NAIR (London, United Kingdom), Yu XIAO, Bastien LECOEUR, Joan CHICK, Sian COOPER, Alison TREE, Petra J VAN HOUDT, Uwe OELFKE, Matthew D BLACKLEDGE, Andreas WETSCHEREK
11:30 - 11:32
#46906 - PG220 Influence of T1-weighted MRI data on convolutional neural network performance on neurodegenerative disease classification.
PG220 Influence of T1-weighted MRI data on convolutional neural network performance on neurodegenerative disease classification.
The use of deep learning has increased significantly in recent years in the field of neuroscience. Various methods are being developed as aid-to-diagnosis tools from imaging data with successful results. Our objective is to study how different parametric maps, all calculated from T1-weighted (T1w) magnetic resonance imaging (MRI), influence the learning process of a 3D convolutional neural network (3D CNN) to classify healthy controls (HC) from patients suffering from multiple system atrophy (MSA), a rare neurodegenerative disease [1].
The dataset comprised T1w MRI acquisitions gathered from three MSA reference centres in France [1-5], including 126 HC and 92 MSA patients. Images were processed using SPM12 on MATLAB R2019b with the following steps: (i) spatial normalization of T1w volumes in the Montreal National Institute (MNI) space (ii) resampling to a 2×2×2 mm3 resolution; (iii) skull stripping; (iv) segmentation of brain substances from the normalized T1w, obtaining grey matter density (GD), white matter density (WD) and cerebrospinal fluid (CSF) maps. We considered a 3D CNN architecture [4] to perform a binary classification task between HC and MSA patients, using a single image type (monomodal) and different combinations from the maps (bimodal, trimodal, and quadrimodal). The CNN was trained for 30 epochs with a ten-time repeated five-folds cross-validation with two different dataset splits. The database was divided into 80%/20% for training/testing our model. We considered accuracy, specificity and sensibility on the hold-out sets to evaluate CNN performance according to the different inputs. In the monomodal study, we obtained the best mean accuracy with GD and WD maps (0.92 ± 0.06 and 0.94 ± 0.06 respectively), followed by T1 and CSF (0.87 ± 0.05 and 0.88 ± 0.04). However, the parametric maps showed a greater difference between sensitivity (0.50-1.00) and specificity (0.89-1.00), compared to the model based on T1 images (T1-CNN). The multimodal study showed an overall improvement in performance, with a mean accuracy of 0.90 ± 0.06 for the T1-GD-CNN and particularly for the T1-GD-WD-CNN, with a mean accuracy of 0.93 ± 0.06. The T1-GD-WD-CSF-CNN yielded results comparable to those of the previously mentioned models with a 0.92 ± 0.06 accuracy. Firstly, the monomodal study enabled us to demonstrate the good classification capabilities of our 3D CNN to distinguish MSA patients from HC. Second, the multimodal study allowed us to observe a general improvement in performance depending on the type of map used with the T1 images. In our bimodal study, we found that adding as input a single parametric map improves the classification capabilities of the 3D CNN, but also can inform about the interpretability of the model from the monomodal performance. For example, considering the T1-GD-CNN, we can suppose that performances will benefit from the better classification found with GD maps alone. In the trimodal approach, we observe an increase in classification capabilities but interpreting these models becomes more challenging as we cannot know to which extent each map contributes to the multimodal performance. Finally, our quadrimodal study yielded classification capabilities comparable to the trimodal study, while increasing computational complexity. It is also important to note that there is a trade-off between the classification capabilities of our model and the computational cost. Indeed, the addition of a modality greatly increases the computational time of the algorithm. Although our sample size is limited by the rarity of the disease, these results are encouraging and show that multimodality can play a role in enhancing diagnostic accuracy. Lastly, to our knowledge, no study has yet exploited the unique contribution of parametric maps derived from the same MRI sequence targeting different aspects of brain structure compared to the sequence itself. This approach could pave the way for a better understanding of the “black box” nature of 3D CNNs thanks to the analysis of input data. However, the use of visualization techniques and the study of misclassified patients could help us to shed light on the impact of parametric maps on the 3D CNN performance. Using different T1-derived maps as input to a 3D CNN increased classification performance to distinguish HC from MSA patients. However, the monomodal study based on the parametric maps calculated from the T1-weighted sequence showed the contribution of each cerebral substance by providing more precise information. To validate our approach, we plan to apply it to other pathologies, such as Alzheimer's disease, and to extend it to the differential diagnosis of parkinsonian syndromes.
Pierre TODESCHINI (Toulouse), Giulia Maria MATTIA, Lydia CHOUGAR, Alexandra FOUBERT-SAMIER, Wassilios G. MEISSNER, Margherita FABBRI, Anne PAVY-LE TRAON, Olivier RASCOL, David GRABLI, Bertrand DEGOS, Nadya PYATIGORSKAYA, Marie VIDAILHET, Jean-Christophe CORVOL, Stéphane LEHÉRICY, Patrice PÉRAN
11:32 - 11:34
#47314 - PG221 Automatic Spinal Cord Gray Matter Segmentation Across Multiple Contrasts, Magnetic Fields, Regions and Pathologies.
PG221 Automatic Spinal Cord Gray Matter Segmentation Across Multiple Contrasts, Magnetic Fields, Regions and Pathologies.
Magnetic resonance imaging (MRI) of the human spinal cord (SC) has seen significant technical advances, enabling the visualization of white matter (WM) and gray matter (GM) across various sequences such as MGE-T2starw, PSIR [1], TSE-T1w, MTR, rAMIRA [2], MP2RAGE [3], and SWI [4], at different field strengths (1.5T, 3T, 7T). GM segmentation is a key step for extracting biomarkers such as cross-sectional area (CSA) [5,6], which are essential for monitoring neurodegenerative and traumatic pathologies including MS [7,8], ALS [2,9,10], SCI [11], SMA [12], PPS [13], and DCM [14]. It also facilitates enhanced image registration, inter-subject alignment within the lumbosacral SC [15], tensor-based morphometry [16], ROI-based qMRI analyses [17], new templates construction [18] and fMRI processing [19–21].
Manual segmentation is time-consuming and subject to inter- and intra-rater variability [15,22,23], prompting the development of automated approaches [5,16,22,24–26]. Among these, the deep learning-based model sct_deepseg_gm [27], integrated into the Spinal Cord Toolbox (SCT) [28], has demonstrated strong performance particularly on MGE-T2starw images and similar contrasts and was the winner of the SC GM Segmentation Challenge [22]. However, the growing diversity of MRI contrasts, acquisition protocols, and use cases, including pediatric and thoraco-lumbar imaging, presents ongoing challenges for model generalization.
This study aims to develop an automatic GM segmentation method that is robust across contrasts, field strengths, SC regions, and pathologies.
Data
This study relies on multicentric data from 16 sites (Figure 1a), including 3 magnetic field strengths and 12 MRI contrasts grouped into 9 categories (Figure 1b). The dataset includes pediatrics controls, adult healthy controls (HC) as well as patients with ALS, MS, SMA, PPS, DCM, SCI, and stroke, for a total of 1,367 volumes (Figure 1c).
Preprocessing
All images were reoriented to RPI orientation and resampled in the axial plane to 0.3x0.3 mm2 using SCT v6.5 [28].
GM Ground Truth
Manual GM segmentations, provided as binary masks, were available from 10 sites. For the remaining 6 sites, GM segmentation was first obtained using a preliminary version of our method (r20250204), followed by manual correction to ensure accuracy.
Automated Segmentation
Benchmark: Contrast-specific models were used as benchmarks (Table 1). For contrasts without existing segmentation methods (PSIR, 3T-TSE-T1w, 7T-MP2RAGE-T1map, and thoraco-lumbar images), single-contrast models were trained using the nnU-Net v2 framework [29].
Our approach: The nnUNetv2 framework was used to train on 1000 epochs in 5 folds with cross-validation on a 2D model.
The training dataset consisted of 1,141 volumes (15,535 axial slices). The testing dataset comprised 20% of the images from sites with GTs (Figure 1a).
Evaluation of Segmentation
Segmentation performance was assessed using the Dice Similarity Coefficient [30] and the Hausdorff Distance (HD) in mm [31], calculated for each 2D axial slice.
Statistics
Wilcoxon test for paired samples, comparing benchmark and our approach. Figure 2 summarizes the Dice scores and HD, showing that our approach significantly outperforms the benchmark methods in multiple sclerosis (MS) cases (p < 0.05). This demonstrates the model’s superior performance in segmenting gray matter in the presence of MS lesions (Figure 3).
A Dice score above 0.85 (Figure 2d) indicates high segmentation fidelity, comparable to that of benchmark #1 [27] reported in the SC GM Segmentation Challenge. However, benchmark #1 shows reduced performance in the lower cervical and thoracic regions (Figure 2a,b). For the 7T datasets, both the benchmarks (#3, #6) and our method performs well. While our method and the benchmarks perform adequately overall, segmentation in patients with SMA and DCM is less accurate (Figure 2a), likely due to SC compression that alters GM shape and contrast. This highlights the need for more training data from varied pathological cases.
In the thoraco-lumbar region (Figure 2b), our model maintains good performance, although segmentation accuracy decreases at the L2 level due to the small size of the spinal cord at this point (~10 pixels; Figure 3). Dice scores are impacted by the limited number of GM voxels in this region across all methods.
Our model achieves an average HD below 0.6 mm across most contrasts, indicating that the predicted GM contours typically deviate by only one pixel from the ground truth. This robustness holds across diverse GM morphologies and pathological conditions (Figure 3) We present a robust approach for automatic GM segmentation, demonstrating consistent performance across multiple MRI contrasts, SC regions, and pathological conditions. Our approach is publicly available in the SCT v7.0.
To further enhance generalizability, especially in challenging pathological cases, future work should incorporate more diverse and pathology-rich datasets during training
Nilser LAINES-MEDINA (Marseille), Jan VALOSEK, Samira MCHINDA, Katerina KREJCI, Josef BEDNARIK, Tomas HORAK, Petr KUDLICKA, Nico PAPINUTTO, Deborah PARETO, Jaume SASTRE-GARRIGA, Alex ROVIRA, Mindy LEVIN, Feroze MOHAMED, Seth SMITH, Tobias GRANBERG, Christopher HEMOND, Charidimos TSAGKAS, Cristina GRANZIERA, Regina SCHLAEGER, Claudia WHEELER-KINGSHOTT, Kristin P. O'GRADY, Gergely DAVID, Virginie CALLOT, Julien COHEN-ADAD
11:34 - 11:36
#45719 - PG222 Performance monitoring of AI-based MR reconstructions via image quality metrics: a two-year study.
PG222 Performance monitoring of AI-based MR reconstructions via image quality metrics: a two-year study.
AI-based MR reconstructions can accelerate scanning and/or improve image quality.
Guidelines for deploying AI tools in radiology[1–4] recommend post-deployment monitoring, as performance may vary due to scanner software updates[5], yet lack specific methods for monitoring that follow established quality control (QC) frameworks: defining performance metrics, baseline values, monitoring frequency, and feedback mechanisms[6–8].
Radiologist assessments are resource-intensive; while phantom-based methods have not been developed for AI-based reconstructions, making both unsuitable for QC. Image quality metrics (IQMs), used to evaluate performance of AI-based images and reconstructions[9–14], offer a more efficient alternative for monitoring.
Our institution introduced an AI-based reconstruction for rectal MRI, evaluated using radiologist scoring[15]. We developed a QC programme using IQMs proven to be sensitive to known perturbations in the AI-based reconstruction[16].
The aim of this study was to evaluate 24 months of QC data, during which the MR systems underwent software upgrades with known modifications to the reconstruction pipeline.
90 patients (47M/43F, 33-89y, median 64y) undergoing routine anorectal MRI (Apr 2023-Apr 2025) were scanned on a 1.5T or 3T system (1.5T-A: n=19, 1.5T-B: n=18, 3T-A: n=36 and 3T-B: n=17, MAGNETOM Sola/Vida, Siemens Healthineers, Forchheim, Germany) using a 30-ch body array and 32-ch spine array. The study was approved by a research ethics committee; patients gave verbal consent for additional imaging as part of routine clinical MRI examinations. Axial T2-weighted small field-of-view turbo spin-echo sequences were acquired with and without AI-based reconstruction[15, 16] at matched positions. Raw data were also saved. Patients were numbered chronologically, the first 50 patients formed the reference dataset; the rest formed the constancy dataset. During the study, the 1.5T-A, 3T-A and 3T-B systems underwent software upgrades.
A radiologist qualitatively reviewed 3 prospective post-upgrade datasets per upgraded system. Post-upgrade reconstructions and difference images were generated for 3 datasets per 3T system (Fig.1).
Paired IQMs – mean squared error (MSE), peak signal-to-noise ratio (pSNR), structural similarity index (SSIM), and visual information fidelity (VIF)[17–19] – and unpaired IQMs – image entropy, and Tenengrad[17, 20] – were evaluated in a 214×214-pixel central region-of-interest for all slices.
IQM values for all datasets were evaluated using Shewhart control charts, comparing each exam's mean IQM against the reference mean and standard deviation (SD). Control chart test rules (Fig.2) were applied[21]. Radiologist scoring was performed for the reference dataset[15].
IQMs were evaluated using MATLAB (R2024b, Natick, MA). Additional QC data acquisition took an average of 6 minutes per exam.
Difference images (Fig.1B) showed changes in accelerated acquisitions with a mean pixel intensity of 0.4 ± 0.6 whereas standard acquisitions showed minimal change, with a mean pixel intensity of 0.0007 ± 0.04.
Mean IQM values for patients 56 (SSIM, VIF), 60 (SSIM) and 85 (MSE, pSNR, SSIM, VIF) exceeded 2 SDs, triggering control chart rule 1 (Figs.2&3). Rules 2-4 were not triggered. Mean IQM values for post-upgrade retrospective reconstructions did not trigger any control chart rules.
Only rule 1 was triggered for patients 56 (SSIM, 1.5T-A); 60 (MSE, pSNR, SSIM 3T-A) and 85 (MSE, pSNR, SSIM, VIF, 1.5T-A) when evaluating the IQMs per scanner (Fig.4). This study evaluated IQMs for QC of AI-based MR reconstructions over 2 years during which there were system software upgrades.
Longitudinal IQM trends indicated stable performance since clinical deployment as no results triggered additional inspection of data or re-evaluation of the imaging.
Examinations with outlier IQM values were traced to gross motion. Motion was generally minimised through patient setup and use of antispasmodics.
No IQM values indicated changes outside of expected variation for examinations conducted post software upgrade, suggesting image quality is consistent with reference data for which radiologist scoring indicated good image quality.
Differences images showed that software upgrades altered the reconstruction pipeline - specifically, a change in the phase correction method for the AI-based reconstruction. Despite this, neither IQMs nor radiologist review indicated changes in image quality.
The IQMs provide a resource-efficient QC method, with automated, quantitative analysis with minimal added acquisition time and no extra radiologist burden. IQMs offer a resource-efficient alternative to routine radiological evaluation for ongoing monitoring of AI-based MR reconstructions. No clinically significant performance changes were detected by IQMs or radiologists after software upgrades. Future work will include radiologist scoring of post-upgrade data and expansion of this QC framework to other MR sequences.
Ciara HARRISON (London, United Kingdom), Owen WHITE, Joshua SHUR, Francesca CASTAGNOLI, Geoff CHARLES-EDWARDS, Brandon WHITCHER, David COLLINS, Matthew CASHMORE, Matt HALL, Spencer THOMAS, Andrew THOMPSON, Georgina HOPKINSON, Dow-Mu KOH, Jessica WINFIELD
|
Salle Major |
|
"Saturday 11 October"
Added to your list of favorites
Deleted from your list of favorites
D32
11:00 - 12:30
FT2-3 - Brain Banks
FT2: Cycle of Translation
11:00 - 12:30
Advancing the diagnosis and treatment of neurological diseases with the help of brain banks.
Helena RADBRUCH (Keynote Speaker, Germany)
11:00 - 12:30
Challenges and pitfalls of setting up MRI protocols for brain banks.
Liana GUERRA SANCHES (Postdoctoral Fellow) (Keynote Speaker, Montreal, Canada)
11:00 - 12:30
The digital brain bank.
Benjamin TENDLER (Keynote Speaker, Oxford, United Kingdom)
|
Salle 120 |
|
"Saturday 11 October"
Added to your list of favorites
Deleted from your list of favorites
E32
11:00 - 12:30
MS2 - Self-learning MRI
|
Salle 76 |
|
"Saturday 11 October"
Added to your list of favorites
Deleted from your list of favorites
G32
11:00 - 12:30
Poster 6
FT2 - Diffusion | FT1 - Acquisition methods | FT1 - Spectroscopy and X-Nuclei | FT2 - Functional and metabolic MRI | FT1 - New technologies for new application
11:00 - 12:30
#47419 - PG465 Comparison of diffusion-weighted imaging for detection of spontaneous muscular activities to clinically established methods: Preliminary results.
PG465 Comparison of diffusion-weighted imaging for detection of spontaneous muscular activities to clinically established methods: Preliminary results.
Diffusion-weighted magnetic resonance imaging (DW-MRI) is able to visualize focal spontaneous muscular activity (SMAM) [1]. This technique is based on the signal reduction induced by the three-dimensional incoherent motion pattern of muscular contraction and has shown a strong correlation to surface electromyographic measurements [2,3]. Furthermore, an increased rate of visible spontaneous activities was found in patients suffering from amyotrophic lateral sclerosis. [3-7] However, a systematic comparison with clinically established methods, e.g., muscle ultrasound and needle electromyography, for the detection of spontaneous activity is still missing.
In this work, the first attempt to compare the results of DW-MRI with two clinically established measurement methods in a cohort of five healthy volunteers is presented.
DW-MRI: Time-series of diffusion-weighted imaging were acquired from five healthy subjects (age: 29.4±4.6 years) on a 3T MR system (MAGNETOM Prismafit, Siemens Healthineers AG, Forchheim, Germany) using a diffusion-weighted stimulated-echo (DW-STE) EPI research sequence. Following muscle groups were examined: tibialis anterior (TA) and gastrocnemius medialis (GM) of the left and right lower leg, paraspinal muscles at the position of the vertebra Th10 (PM), deltoid muscles (DM) and tongue muscles (TM). Protocol parameters of the DW-STE-EPI were chosen according to Schwartz et al. [4] and are given in Table 1. 500 repetitions were chosen leading to a total scan-time of 250 s for each muscle group.
Muscle ultrasound (US): For comparison, muscle ultrasound images were acquired at all aforementioned muscular regions with an examination time of 120 s per muscle using a Canon Aplio i800 ultrasound system.
Needle electromyography (nEMG): Due to the invasiveness of this measurement technique, examination was restricted to the TA and GM of the two lower legs. In each case, 3 needle positions were examined along the muscle with an overall acquisition time of 90 s (30 s per needle position). A Sierra Summit (Cadwell, USA) system was used for recording the myoelectric signals.
The study protocol was approved by the local ethics review board of the medical faculty of the Eberhard Karls University and the University Hospital of Tübingen in 830/2020BO2.
Evaluation: DW images were intra-subject registered before applying a neural network approach [5] for detection and segmentation of spontaneous muscular activities. All segmentation results were manually revised to ensure proper classification and were visualized as percentage Event Count Maps (pECM), i.e., the summation of events in time direction normalized by the number of repetitions, using custom-built tools in MATLAB® (The MathWorks, Natick, MA, USA). Spontaneous activities in muscle ultrasound images were visually detected by an experienced specialist during the examination. Results of the nEMG measurements were also manually counted. All results were reported as rate of activity per minute. Relation between nEMG vs. DW-MRI and US vs. DW-MRI was examined with a Spearman correlation. Exemplary pECMs of Subject #4 (highest mean rate) for each individual muscle group is depicted in Figure 1. It can be seen that an overall high rate of activity is detectable in the GM of the right and left lower leg. This is in good accordance to the detected activity rate of nEMG and US. The rate of activity for each modality and muscle group is given in Table 2. A significant correlation for nEMG (ρ = 0.64, p = 0.003) and US (ρ = 0.49, p = 0.0008) was found. In two subjects, the tongue muscle could not be analyzed due to non-resting musculature and enhanced imaging artifacts caused by a dental retainer. There is a significant agreement between the various muscle groups and modalities. However, it must be noted that in contrast to US and nEMG, DW-MRI was evaluated on the entire cross-sectional area of the muscle instead of a more localized area. Depending on the parameterization, there is a certain probability of missing a spontaneous activity in DW-MRI, e.g., start of muscle contraction after image readout [8]. These two influencing factors must be investigated further. Recording and evaluating the tongue muscles appears to be more difficult, since this muscle region is not always in a resting posture, depending on the subject. The detection results of DW-MRI show good agreement with clinically established methods (US and nEMG) in a small cohort of healthy volunteers. Further studies are needed to investigate the correlations between all three modalities in a patient cohort.
Martin SCHWARTZ (Tuebingen, Germany), Petros MARTIROSIAN, Julia WITTLINGER, Thorsten FEIWEIER, Guenter STEIDLE, Bin YANG, Ludger SCHÖLS, Fritz SCHICK
11:00 - 12:30
#47560 - PG466 Frequency-dependent diffusion tensor distribution imaging in the evaluation of ischemic stroke.
PG466 Frequency-dependent diffusion tensor distribution imaging in the evaluation of ischemic stroke.
Ischemic stroke is a global health and economic burden, with up to 7.6 million incident cases annually [1]. The imaging techniques used in clinics effectively discern large-scale structural damage in the core, however, they often fail to detect subtle changes and estimate tissue viability in the lesion, penumbra and distal regions. [2-4]. Brain imaging urgently requires solutions.
Frequency-dependent diffusion tensor distribution imaging (ωDTD) is an emerging method that exploits frequency-dependent tensor-valued diffusion encoding for sub-voxel parameter estimation [5,6]. To extract within the voxel information on signal-fractions specific to certain tissue characteristics we exploit gaussian mixture-model based clustering of tensor-valued distributions as in [7] as opposed to traditional, manual bin-segmentation [6].
Here we have combined ωDTD, clustering of the per-voxel parameter distributions and multivariate statistical modeling to capture the relationship between MRI and histology following an ischemic stroke.
Eleven 4-month-old male Wistar Han rats 250-450 g, were used in this study. A transient ischemic injury was induced in the left hemisphere using the intraluminal middle cerebral artery occlusion model described in [8,9]. The animals were transcardially perfused at 24-hours post-reperfusion.
Ex vivo MRI was acquired in a 11.7-T Bruker Avance-III HD spectrometer in a 10-mm volumetric coil using a multi-slice multi-echo 2D sequence customized for tensor-valued diffusion encoding. The sequence included variable b-values (700 - 8000 s/mm^2), normalized anisotropy b∆ (-0.5, 0, 0.5 and 1), TR = 800 ms, TE = 30 ms, orientation (θ, ϕ), centroid frequency ω_cent/2π = 34 - 115 Hz, and an in-plane voxel resolution of 80 x 80 μm^2. After, the brain sections were prepared for Nissl staining. The clustering of diffusion tensor distributions was performed as in [7].
For the regression analysis (Fig.1.), quantitative histological parameters were extracted from the Nissl micrographs using QuPath. These parameter maps were downsampled and registered with MRI. Both MRI and histological parameter maps were preprocessed and entered into the random forest (RF) model. The performance of the RF [10] was evaluated with cross-validation leave-one-animal-out (CV LOO) and common metrics such as R^2, R, Q^2, MSE and MAE. Additionally, we investigated how the prediction accuracy depends on the input MRI parameters, by training a separate RF model with both conventional diffusion tensor imaging (DTI) and three different ωDTD parameter sets (per-voxel means, bin-resolved and cluster-resolved fraction maps). Fig.2. shows how the RF predictions depend on the input MRI parameters. The predicted number of cells based on conventional MRI (Fig.2. D) shows a scenario where the regression model has failed to capture the more nuanced tissue changes. However, the predicted number of cells based on bin-resolved ωDTD (Fig.2. E) resembles more closely the original histological parameter map both in lesion details and values.
The original histological parameters and their counterpart predictions based on ωDTD cluster-resolved fractions are shown in Fig.3. The predicted maps replicated the distinct lesion boundaries and characteristics of each histological parameter (white arrows), such as the layer of increased number of cells in the cortex. The performance metrics for evaluating the RF are shown in Table.1. We found that the ωDTD parameters were highly predictive of changes in cellularity and morphology of cells, including alterations in nucleus size and shape. This indicates that the ωDTD parameters are more descriptive of the histological changes, i.e. more strongly related to the target variable. The R^2 values showed our models explained between 35% - 70% of the variance present in the data. The correlation R was also consistently positive for the CV, showing that our models repeatedly captured a relationship between the input MRI parameters and the target histological parameters. The variability observed in both MSE and MAE across CV folds suggests inter-subject differences. In the future, additional histological stains and expansion to fully 3D analyses incorporating deep learning algorithms could open new perspectives in predicting tissue changes. In conclusion, ωDTD can open new avenues in the evaluation of ischemic stroke by capturing subtle cellular-level alterations in tissue viability, composition, and microstructure.
Sara GRÖHN (Kuopio, Finland), Angela NARANJO, Omar NARVAEZ, Maxime YON, Buse BUZ-YALUG, Santos BLANCO, Daniel TOPGAARD, Esther MARTINEZ-LARA, Ma Angeles PEINADO, Jussi TOHKA, Alejandra SIERRA
11:00 - 12:30
#47606 - PG467 Reaching the Tail: Validating MRI Axon Radius Mapping with MRI-Scale Histology.
PG467 Reaching the Tail: Validating MRI Axon Radius Mapping with MRI-Scale Histology.
The axon radius is a promising clinical biomarker for neurological disorders and may be accessible via non-invasive diffusion-weighted MRI (dMRI). One candidate dMRI model estimates the effective axon radius
r_eff = (⟨r^6⟩ / ⟨r^2⟩)^(1/4),
which emphasizes the tail of the axon radius distribution [1-2].
However, r_eff remains insufficiently validated against ex vivo histology. While qualitative comparisons hint at common spatial patterns in the corpus callosum [3-9], there is no quantitative proof that r_eff reflects tissue microstructure. This gap is due to limited variation [2] or sparse sampling [3,8,10-16] in existing histology. Additionally, the small ROI sizes likely yield imprecise r_eff - due to its tail-weighting [2,11].
Here, we quantitatively validate r_eff via spatial correlations between in vivo and ex vivo dMRI and corresponding histology, using 35 in vivo dMRI voxel-sized ROIs from two human corpora callosa.
We acquired light microscopy (LM) images for 35 ROIs of two human corpus callosum samples (Fig. 1a–b). We segmented axons [17] per ROI and estimated r_eff. For in vivo dMRI, we acquired magnitude data of five healthy adults (age: 31 ± 3 years; sex: 2 male, 3 female) on a Siemens Connectom 3T scanner at MPI-CBS, Leipzig, Germany, following the protocol in [4]. Briefly, we applied b = [0.5 1, 2.5, 6, 30.45] ms/µm^2 for [30, 30, 30, 60, 120] gradient directions with variable gradient amplitude and 2.5 mm isotropic resolution. We also acquired T1-weighted MPRAGE and non-diffusion-weighted images for geometric susceptibility correction. We corrected for Gibbs ringing artifacts [18-19], geometric susceptibility, eddy currents, motion [19-21] and gradient distortions [22-23]. For b ≥ 6 ms/μm^2, we estimated powder-averaged signals per b using a Rician maximum likelihood (ML) estimator [24] relying on a noise level estimate [19], [25], [26]; subsequently, we estimated r_eff [5]. For b ≤ 2.5 ms/μm^2, we mapped DKI-based fractional anisotropy (FA) [19,27-28]. For ex vivo dMRI, we acquired magnitude data using a Bruker Biospin 9.4T scanner at the Berlin Ultrahigh Field Facility in Berlin, Germany, using a protocol akin to [2]. Briefly, we used a segmented EPI sequence with b = [20, 30, 40, 50, 60, 70, 80, 90, 100] ms/µm^2 for 65 gradient directions per b with variable gradient amplitude and 0.35 mm isotropic resolution. To enhance SNR, we averaged up to 8 images for high b. We corrected for Gibbs ringing artifacts using MRtrix3 [18], [19]. We estimated the powder-average per b using a Rician maximum likelihood (ML) estimator [24]. We subtracted the immobile water compartment signal [29], estimated from signals at b = 100 ms/µm^2 with strong alignment to the main fiber direction. Finally, we estimated r_eff [30]. For qualitative dMRI-histology comparison, we assessed spatial r_eff patterns for all modalities in the mid-sagittal slice of MNI space (see Fig. 2 for registration). For quantitative comparison, we assessed absolute agreement across ROIs evaluated in their native spaces using the normalized root-mean-square error (NRMSE), normalized by the mean of histological r_eff (Fig. 2). We assessed sensitivity via Pearson’s R and its two-sided p-value using Monte Carlo permutation with K = 10⁶ permutations (null hypothesis: R = 0). Fig. 3 compares group-level in vivo dMRI-based r_eff with histology. The spatial patterns (Fig. 3a) appear slightly overestimated and show reduced dynamic range, suggesting limited microstructural sensitivity. Still, dMRI captures a coarse histological trend, with alternating low–high r_eff across anterior midbody, midbody, posterior midbody, and splenium. The quantitative analysis (Fig. 3b) confirms this agreement via significant correlation (R = 0.41, p = 0.019).
Fig. 4 show the ex vivo counterpart to Fig. 3 based on a single sample (CC-01). The spatial pattern (Fig. 4a) shows low dynamic range and no clear structure, resulting in the absence of significant correlation (Fig. 4b, R = 0.23, p = 0.41). While the strong dynamic range reduction may be inherent to ex vivo dMRI and tissue, it may also reflect the added model complexity introduced by the immobile water compartment. We provide the first quantitative evidence that r_eff, and axon morphology more broadly, are detectable in human brain in vivo dMRI. This is despite recent 3D histology revealing complex axonal morphology [12,14,16,31-32] which challenges the perfect cylinder assumption of r_eff. Consequently, 2D histology emerges as a scalable method for experimental validation, complementing simulations based on sparse 3D axonal reconstructions. The low ex-vivo sensitivity suggests that using ex-vivo dMRI as an intermediate validation step to bypass inter-individual differences may be inherently difficult. Our demonstration of dMRI’s sensitivity to axon morphology motivates advances to address the observed dynamic range reduction, better model realistic axonal morphology, and support clinical translation.
Laurin MORDHORST (Luebeck, Germany), Luke J. EDWARDS, Maria MOROZOVA, Mohammad ASHTARAYEH, Tobias STREUBEL, Björn FRICKE, Francisco J. FRITZ, Henriette RUSCH, Carsten JÄGER, Starke LUDGER, Thomas GLADYTZ, Ehsan TASBIHI, Joao S. PERIQUITO, Andreas POHLMANN, Thoralf NIENDORF, Nikolaus WEISKOPF, Markus MORAWSKI, Siawoosh MOHAMMADI
11:00 - 12:30
#47708 - PG468 Using Mahalanobis Distance to Detect Along-Tract DTI Abnormalities Predictive of Long-Term Outcome After Traumatic Brain Injury.
PG468 Using Mahalanobis Distance to Detect Along-Tract DTI Abnormalities Predictive of Long-Term Outcome After Traumatic Brain Injury.
Traumatic brain injury (TBI) is a leading cause of morbidity and mortality in individuals under 45 years of age [1-3]. Despite this, our ability to predict functional outcomes following injury remains limited.
Accumulating studies have highlighted the utility of diffusion tensor imaging (DTI) in aiding outcome predictions through its sensitivity to traumatic axonal injury, a hallmark histopathological feature of TBI [4,5]. However, a common limitation of such studies is the reliance on whole-tract averages, which may obscure small but still clinically relevant injuries. Alternatively, these studies rely on voxel-wise group comparisons, which assume consistent injury patterns across the patient cohort. Given the substantial heterogeneity of mechanisms in which TBI can be sustained, this assumption may not be warranted.
Hence, analysis methods that can capture subtle abnormalities across white matter tracts, without requiring spatial overlap of injuries across individuals are needed. One such approach is using Mahalanobis distance, a multivariate outlier detection method that quantifies the extent to which there are regions along the tract that deviate significantly from the reference group. By applying this method to DTI metrics sampled from multiple points along a given tract, it becomes possible to identify subject-level abnormalities that might otherwise be overlooked.
Here, we adapted a previous approach [6] by applying a robust version of Mahalanobis distance, using the minimum covariance determinant [9,10], to quantify abnormalities in axial diffusivity (AD) and radial diffusivity (RD) along several white matter tracts. The study cohort consisted of 18 adult TBI patients (mild to severe) and 18 healthy controls from a previous study [7]. Diffusion-weighted images were acquired on a 3T Siemens Magnetom Verio scanner at the Oxford Acute Vascular Imaging Centre, along 64 directions (b=1500s/mm²) with 5 b=0 images (2×2×2mm voxels, TR=2153ms, TE=85ms, MB=2). Two datasets with opposing phase-encoding directions (A-P, P-A) were collected for distortion correction. Patients were scanned within 24 hours post-injury, and 17 re-scanned at 7–15 days, whilst controls were scanned once.
An automated tractography pipeline (pyAFQ [8]) was used to reconstruct 28 white matter tracts per subject, sampling 100 equidistant nodes along each tract to generate tract profiles. To ensure tract reliability and data completeness, only tracts with >10 streamlines in all participants were retained, resulting in 25 tracts for analysis. For each tract, mean AD and RD values were extracted at all 100 nodes. Robust Mahalanobis distance was calculated for each subject by individually comparing their entire AD and RD tract profiles to the healthy control group, with larger distances indicating a greater deviation from normal.
For each tract, correlations between AD and RD Mahalanobis distances and functional outcome, measured by the Glasgow Outcome Scale–Extended (GOSE) [11] at 6 months post-injury (range: 1 = dead to 8 = upper good recovery), were assessed using Spearman’s rank correlation, with p-values adjusted for a 5% false discovery rate. Within 24-hours post-injury, greater AD Mahalanobis distance in the right anterior thalamic radiation was significantly associated with a lower GOSE score at 6 months (Spearman ρ= –0.82, p=0.0015). However, at 7–15 days post-injury, only increased AD Mahalanobis distance in the right inferior longitudinal fasciculus (ρ= –0.75, p=0.013) and in callosal fibres projecting to the superior parietal lobule (ρ= –0.82, p=0.0031) were significantly correlated with poorer outcome. No other significant associations were identified. Our findings demonstrate that robust Mahalanobis distance applied to along-tract DTI profiles may be able to identify clinically meaningful abnormalities that predict long-term functional outcome in TBI patients. Unlike the typical approaches that rely on tract-averaged metrics or voxel-wise group comparisons, Mahalanobis distance provides a subject-specific, multivariate assessment of the magnitude of abnormality across the whole tract.
Interestingly, the predictive tracts differed between the first and second timepoints, potentially reflecting the evolving nature of TBIs. In a mixed-severity cohort, patients may recover or deteriorate at varying rates, which in a small sample may have reduced the stability of group-level effects. Our findings also suggest that AD may be more sensitive to outcome-relevant axonal abnormalities in the early post-injury stage, compared to RD. Nevertheless, this study is limited by the lack of covariate adjustment (e.g., for age, sex, or lesion burden). Mahalanobis distance may be a promising approach for detecting along-tract abnormalities and aiding outcome predictions following TBI. Future work should implement multivariate outlier detection methods in larger, covariate-controlled cohorts to further assess their utility.
Izabelle LÖVGREN (Oxford, United Kingdom), Michiel COTTAAR, Natalie VOETS, Tim LAWRENCE
11:00 - 12:30
#47800 - PG469 Cerebral Magnetic Resonance Imaging Insights into Bariatric Surgery-induced Changes in Obese Mice.
PG469 Cerebral Magnetic Resonance Imaging Insights into Bariatric Surgery-induced Changes in Obese Mice.
Obesity is a chronic disease associated with several pathologies such as type 2 diabetes, cardiovascular risk or neurodegeneration. Bariatric surgery (BS), initially a high-risk weight-loss procedure, is still the most successful approach at restoring non-obese body mass indexes (BMI) in an increasing range of population [1]. Beyond weight reduction, BS alters metabolism, gut microbiota, and brain function. [2,3]. Among available techniques, vertical Sleeve Gastrectomy (VSG) is one of the most common, removing 70%–80% of the stomach. This study aims to investigate VSG's effects in the brain on a rodent diet-induced obesity model using diffusion tensor imaging (DTI) and magnetization transfer imaging (MTI).
26 C57BL/6 mice (male and female) were fed with a high-fat high-sugar (HFHS, 45kcal% fat, 30 kcal% sucrose) diet (Research Diets Inc., D08112601i). After 20 weeks, obese mice were assigned to either a sham-operated or BS group. Post-surgery, animals were maintained on a liquid diet for 1 week, followed by a chow diet for 6 weeks. Body weight (BW) was followed daily. Brain diffusion tensor images (DTI, 30 directions, TR/TE= 3000/37.56 ms, δ/Δ= 4/25ms, Mtx= 128×128, slice thickness= 1.25 mm, b-values= 800µm2 /s & 2500µm2 /s) and magnetization transfer images (MTI, MTON/OFF, TR=2500ms, TE=9.8ms, and Av=1) were acquired pre-surgery and at 3- and 6-weeks post-surgery using a Bruker Biospec 7T system (Bruker Biospin, Ettlingen, DE). Image pre- and processing was performed using a software based on Dipy [4]. Parametric maps of mean, axial and radial diffusivities (MD, AD, RD), anisotropic fraction (FA) and MT ratio (MTR) were obtained, and regions of interest (ROI) from the hypothalamus (Hyp), hippocampus (Hipp), nucleus accumbens (NAc), and infralimbic area (ILA) selected. Statistical analyses were performed using R [5], and MR parameters were fitted to a variety of LME regression models using the “lme” function of “nlme” [6] package to evaluate the differences of the MR parameters between sham and BS groups over time. Correction for multiple comparisons accross the tests on MRI parameters was achieved by the hierarchical Benjamini-Hochberg procedure [8] using the structSSI package [9]. Briefly, such approach levels groups of hypotheses hierarchically, and only if superior null hypothesis are rejected, the subsequent are tested. BS was associated with a significant reduction in body weight, with mean values of 43.11 ± 6.12 g at pre-surgery, 31.54 ± 3.40 g at 3-weeks post-surgery, and 32.98 ± 3.31 g at 6-weeks post-surgery. The LME analysis of MRI parameters in Hyp revealed significant interactions between “Type” (Sham vs. BS) and “State” (Pre, Post3w, or Post6w) for RD, MD, and FA values (p < 0.001, p < 0.001, and p < 0.05, respectively). Post-hoc tests indicated a significant increase in RD values in the BS group from both Pre (p<0.01) and Post3w (p<0.05) to Post6w, while the Sham group showed a significantly decrease in RD from Post3w to Post6w (p<0.01) (Figure A). Similarly, MD values in the BS group increased significantly from Pre to Post6w (p<0.05) (Figure B). Additionally, group comparisons at each time point confirmed that by week 6, the BS group exhibited significantly lower FA values (Figure C) and higher MD and RD values compared to the sham group (p<0.05 for all metrics). Our results suggest that BS induces a reduction in body weight and distinct and time-dependent changes in MRI parameters. By week 6, the BS group exhibited higher RD and MD values, along with lower FA values compared to the sham group. These findings are consistent with a successful reversal of the obesity-induced inflammatory state, as the observed increases in RD and MD may reflect an expansion of the extracellular space or alterations in cellularity. [7]. We found longitudinal changes in DTI parameters following bariatric surgery, with a reduction in body weight, demonstrating that the effects of bariatric surgery on the brain can be detected in vivo using MRI. Currently, our dataset comprises only n = 13 with the three time points. Future research will increase the sample size and extend the analysis to additional brain regions.
Adriana FERREIRO (Madrid, Spain), Maya HOLGADO, Raquel GONZÁLEZ-ALDAY, Pilar LÓPEZ-LARRUBIA, Blanca LIZARBE
11:00 - 12:30
#47355 - PG470 Adaptive prescan calibration routines and connection to the gammaSTAR framework for accurate sample probing in tabletop MRI system.
PG470 Adaptive prescan calibration routines and connection to the gammaSTAR framework for accurate sample probing in tabletop MRI system.
The rising interest in low field MRI (<1.5T) is uniting a community that has the potential to make a meaningful impact in this niche area of MRI. We here investigate the use of the commercially available Open-Source Console for Realtime Acquisition (OCRA) tabletop system[1] to expand the range of application of portable low-field systems by allowing a connection with gammaSTAR[2], a scanner-agnostic MRI pulse sequence development framework. Previous work [3] has presented a wrapper software that resolves differences between OCRA machine code and the open-source pulse sequence programming environment PulSeq, using MAgnetic Resonance COntrol System (MaRCoS[5,6]) server. In this work, we developed a driver that translates gammaSTAR sequences into the input data format for the MaRCoS server. The connection to the gammaSTAR backend is direct, enabling on-the-fly generation of MRI sequences and flexible changes of sequence protocols. The entire pipeline encompasses a dynamic system calibration workflow, acquisition data storage in the ISMRM Raw Data format [7], and image reconstruction performed using gammaSTAR reconstruction server.
The original Red Pitaya 125-14 in the OCRA console was substituted with Red Pitaya 122-16[8]. A gammaSTAR software driver was implemented to convert gammaSTAR sequence raw data format from the interpreter to MaRCoS numpy arrays input data. The wrapper rearranges the gammaSTAR sequence information into numpy arrays of times and amplitudes for each hardware output and control (radiofrequency pulses, gradient pulses, along with adc data) to be fed as input to the MaRCoS server. Prior to data acquisition, calibration measurements were carried out through the implementation of a calibration routine [1] utilizing a gammaSTAR 1D spin echo sequence for the Larmor frequency sweeping and a 1D spin echo with slice selection sequence for the gradient shimming. Tests were performed using 3D printed shaped cylindrical phantoms (8 mm diameter and 30 mm length) with tap water mixed with copper sulfate (phantom A), and only water (phantom B), along with a plant stem (9 mm diameter and 28 mm length of pseudobulbs of noble dendrobium). The execution of customized pulse sequences was performed by using an acquisition pipeline in python for sending, receiving, and reconstruction. Acquired data were streamed to the gammaSTAR reconstruction server which returns final images in DICOM format. Different 2D and 3D sequences were tested: rapid acquisition with refocused echoes (RARE), balanced steady-state free precession (bSSFP), single shot spin echo echo-planar imaging (SE EPI) and fast low-angle shot (FLASH). Calibration measurements enabled the acquisition of images with a 30 parts-per-million (ppm) B0 inhomogeneity at a Larmor frequency of 11.278 MHz. Figure 1 illustrates the workflow of the connection between gammaSTAR frontend and MaRCoS server for executing user-defined sequences, incorporating both a dynamic calibration pipeline using an iterative method to determine the centered Larmor frequency, gradient shimming offsets, and a data acquisition pipeline. Table 1 summarizes the sequences imaging parameters employed across different phantoms/materials. Figure 3(a-d) illustrates different 2D sequences with very high signal-to-noise (SNR) ratio, including more challenging sequence such as SE-EPI with phase correction. Figure 3e shows a 3D FLASH sequence of a pseudobulb of noble dendrobium specimen with a short echo time, enabling the imaging of materials with low water content, primarily consisting of alkaline substances. We have developed a driver that links the gammaSTAR platform to the OCRA tabletop system through the MaRCoS server, allowing for the design of sequences with various features. We anticipate that this approach will enable efficient validation of MRI sequences on tabletop systems, with direct transfer to full-scale scanners without modification, facilitated by the translational compatibility of gammaSTAR. This would facilitate a quicker and more efficient development of customized sequences. Furthermore, a range of pulse sequences implemented on a clinical MRI scanner have demonstrated robust performance across diverse sample probes on a low-field system, supporting their potential for broader implementation in advanced applications, such as materials science. Additionally, for fast sequences such EPI, further validation of distortion and phase correction techniques may be required for optimal performance. We established a technically functional workflow for OCRA low-field MRI scanner, demonstrating the deployment of optimized custom sequences executed using gammaSTAR and the MaRCoS server. Furthermore we presented illustrative use cases that highlight the versatility of different sequence types moving beyond the hardcoded sequences used in previous work. This establishes a foundation for developing new sequences and exploring innovative applications in scientific research.
Juela CUFE (Bremen, Germany), Daniel Christopher HOINKISS, Simon KONSTANDIN, Jörn HUBER, Matthias GÜNTHER
11:00 - 12:30
#47609 - PG471 Detection and parsing of MRI sequences using an autonomous field camera.
PG471 Detection and parsing of MRI sequences using an autonomous field camera.
Measuring and characterizing gradient fields in MR sequences using NMR field probes is a frequently recurring aspect to for example modern image reconstruction, sequence development and sequence evaluation. In particular, the evaluation and testing of sequences can be a powerful tool in debugging erroneous gradient pulses or in understanding the fundamental composition of a sequence. Such measurements can however normally only be made if the user has special access rights to the scanner platform and if they can incorporate trigger pulses and probe position calibration sequences onto the scanner. In this abstract, we show how ‘true’ sequence diagrams can be characterized, including their imperfections, for unknown sequences using a scanner independent field camera.
Our proposed system employs a clock that is fully separated from the scanner and uses software-driven triggering for probe measurements, illustrated in Fig. 1 [1]. By relying on the periodic nature of MR sequences, the system timing can be aligned with that of the sequence by determining the repetition rate. Once the repetition rate is found, pseudo-continuous measurements of the TR can be provided which can serve as a basis for position calibration, Fig. 2.
To turn field measurement into measurement of gradients, the probe positions need to be calibrated. To do this, the probes are set up in pairs with known distances, where the method presented in Fig. 3 is applied to find the probe positions in the imaging coordinate system. This method is compatible with a large set of different sequences and does not rely on knowing the exact gradients beforehand.
To produce the final gradient waveforms illustrated in Fig. 3 we however need to overcome two obstacles: First, they have some unknown individual offsets O, and are in the intermediate step in Fig. 3 also rotated by some unknown 3D rotation matrix. The offsets occur due to individual probe off-resonances, illustrated in the top right of Fig. 3, caused by local field inhomogeneities across each probe. The determination of these offsets can be done in a few different ways: The simplest is to directly measure them while the scanner is inactive and use these values for all subsequent measurements. The disadvantage with this approach is that if the scanner applies shimming gradients, or if the probes are displaced, these values may become obsolete. A different method that accounts for this is to ensure that the sequence used for calibration features some gradient ‘silence’ and use those windows for explicit measurement of the offsets in real time.
The missing rotation stems from the step in the calibration method that employs the known calibrated rigid body constraints: These will generally have been determined in some arbitrary orientation which does not match the present one. Correcting for this rotation can be done by identifying some isolated and mutually orthogonal gradient segments, such as readout and slice select, both of which are generally found in cartesian imaging sequences.
Going through the steps above, a set of sequence diagram-style waveforms can be produced, without the probes ever needing any scanner input. For the sake of creating an undistorted sequence diagram, the imaging RF was disabled during measurements. A set of calculated sequence diagram waveforms together with their corresponding reference waveforms are illustrated in Fig. 4. The full calibration procedure has been applied for each of these three sequences, which means that they are all compatible with our method and can be used to calculate the probe position. Because the phase encoding waveforms change throughout the sequence, the measurements have been scaled to match the reference waveforms. The agreement between measurement and reference suggests that the position calibration method has successfully determined the probe positions, which in turn translates into precise measurements of gradient strengths. In this work we show how to parse sequence diagrams of unknown sequences using an autonomous field camera. Our method does expect some basic features in the sequence being measured, such as it having gradient silence or it using cartesian imaging. This does rule out a subset of sequences from direct use for calibration, but still leaves the possibility of using any compatible sequence for initial position calibration and later reusing that same calibration for fully arbitrary sequences.
Producing sequence diagram results as we have done in this abstract has potential in sequence development, hardware diagnostics and in understanding the exact physical implementation of sequences, and can further be used on scanners that do not have the appropriate hardware and software integrated for regular field camera measurements. This could also be useful for MR sites that do not have research agreements with their vendors, or for mobile field camera systems that could be readily moved between different scanners and scanner platforms.
Oskar BJORKQVIST (ZURICH, Switzerland), Klaas P. PRUESSMANN
11:00 - 12:30
#47595 - PG472 The benefits of magnetic resonance simulation for emerging technologies: an emblematic application to magnetic-resonance-guided gamma imaging.
PG472 The benefits of magnetic resonance simulation for emerging technologies: an emblematic application to magnetic-resonance-guided gamma imaging.
Magnetic resonance (MR) simulation is a key enabler in exploring unconventional imaging approaches where no physical device yet exists. Magnetic-resonance-guided gamma imaging (MRG-γ)—based on the anisotropic γ-emission of hyperpolarized metastable nuclei controlled by MR manipulation of spin—is a promising but experimentally inaccessible modality [1–3]. Therefore, realistic simulation is crucial for validating physical models, characterizing parameters, and informing the design of acquisition strategies and hardware architecture. Although recent MR simulators have expanded their support for quantitative imaging, artificial intelligence, and sequence prototyping, they are not suited to model spin behavior at the γ-event level or to simulate γ image formation [4–9]. Our simulator, MRGS, addresses this gap by integrating spin dynamics, anisotropic γ-emission modeling, γ-detection, and γ-image reconstruction, enabling in-silico MRG-γ. This work illustrates the broader value of MR simulation in the development of emerging MR techniques.
We developed a modular MRG-γ simulator (see Figure 1) composed of MR and γ simulation parts. The simulation begins with user-defined inputs specifying the MR and γ physical parameters, phantom geometry, and the MR sequence. The simulator first computes magnetization evolution by solving the Bloch equations voxel-wise with high temporal resolution. These spin evolutions are used to stochastically model anisotropic γ-emission based on spin orientation, followed by γ-detection, and then γ-image reconstruction using list-mode maximum likelihood expectation maximization (MLEM) [10–13]. The outputs include magnetization evolutions, detected γ-events, and reconstructed γ images. The design emphasizes temporal fidelity and physical accuracy over spatial resolution and speed, making it uniquely suited for event-scale MRG-γ.
MRGS considers MR and γ parameters, as well as phantom geometries and any MR sequence defined using PyPulseq [14] (see Table 1 for detailed MRG-γ parameters). The MR part numerically solves the Bloch equations using a fifth-order Runge-Kutta method [15, 16] with a time step ranging from 1 to 200 µs, chosen to approach the γ-event scale. The γ-emission and detection are modeled using a probabilistic approach based on Poisson-distributed decay events and voxel-wise angular probabilities defined by P(θ)=a0+a2cos(2θ), where a0 is the isotropic baseline, a2 modulates the emission anisotropy relative to the spin orientation, and θ is the angle between the spin and the detector (see Figure 1 γ-detection) [2, 3]. Detector sensitivity is incorporated through precomputed voxel-to-detector visibility maps, which account for geometric coverage and solid angles (see Figure 1 γ-reconstruction). These maps modulate the emission probability to yield physically consistent detection events. MRGS was evaluated on 2D phantoms under multiple configurations (see last column of Table 1 for tested values). The γ-simulation reproduced expected angular distributions consistent with P(θ)=a0+a2cos(2θ), validating the physical emission model (see Figure 1 γ-emission and γ signal). It also enabled the reconstruction of MR parameters from the γ signal (see Figure 2a). Using a “border” gradient trajectory, we achieved high phase dispersion across voxels (see Figure 2b). This sequence maximized angular discrimination for γ-emission. Simulations confirmed the importance of T₁ and T₂*: shorter T₁ reduced signal amplitude over time, while shorter T₂* increased phase dispersion within voxels, degrading emission directionality and increasing uncertainty in γ-origin. We used isochromats per voxel to assess this effect [17, 18] (see Figure 2c). MLEM reconstruction yielded γ-images that qualitatively matched the underlying source distribution (see Figure 1 γ image). MRGS enabled the design of MR sequences optimized for MRG-γ and the reconstruction of physical parameters (T₁, T₂, a₂) from simulated γ-detection. It achieved high temporal precision, allowing realistic modelling of anisotropic γ-emission. While spatial resolution is limited and full Monte Carlo simulation is avoided, this tradeoff ensures computational efficiency and physical consistency.
Simulations highlight the critical impact of T₂* on localization uncertainty. Excess dephasing reduces angular precision, increasing the required number of detected events. Achieving usable SNR thus depends on longer T₁, T₂, higher polarization, or higher activity. Despite current limitations, the simulator offers valuable insights to guide future MRG-γ designs and acquisition strategies. We presented MRGS, a dedicated MRG-γ simulation framework. By combining spin dynamics with anisotropic γ-emission modeling and image reconstruction, our simulator enables exploration of this novel modality. It supports sequence design, parameter validation, and performance estimation. This work highlights the pivotal role of simulation in the development of emerging MR techniques.
Christophe CHÊNES (Geneva, Switzerland), Pablo GALVE, Marie-Anaïs PETIT, Joaquín LOPEZ HERRAIZ
11:00 - 12:30
#47378 - PG473 MR Spectroscopy without water suppression using the Gradient Impulse Response Function.
PG473 MR Spectroscopy without water suppression using the Gradient Impulse Response Function.
Water suppression in MRS is considered essential due to artefactual sidebands on the water peak, which obscure the metabolite signals. These sidebands arise from field perturbations caused by gradient induced eddy currents in the MRI hardware. However, MRS without water suppression is desirable, as the high signal water can aid in data correction, concentration referencing, and lowering SAR load. Additionally, downfield labile peaks will remain visible without water suppression. Previous attempts[1,2] to achieve MRS without water suppression have fallen short due to hardware limitations and a lack of generalisation. In this study, the effect of system imperfections (eddy currents) are removed by accurately characterising them using the Gradient Impulse Response Function (GIRF) [3]. We implement a post-processing method using the GIRF to correct the artefactual sidebands on the water peak, achieving a generalisable method for non-water suppressed single-voxel spectroscopy (SVS) without requiring additional hardware.
SVS was performed in a 3T Siemens Prisma scanner using the vendor harmonised SEMI-LASER sequence from the CMRR spectroscopy package[4] (20 mm isotropic voxel, TE: 36 ms, bandwidth: 6000 Hz, GOIA-WURST pulses). Non-water suppressed spectra were acquired by disabling the VAPOR water suppression. Water suppressed spectra were acquired as a reference. The study was performed using a SPECTRE phantom (Gold Standard Phantoms, Sheffield, UK), with voxel locations at isocenter and 20 mm offsets; and in one healthy participant with voxel locations at isocenter and the primary motor cortex.
The GIRF was measured in the same scanner following the optimised protocol established by Wu[5]. Additional 5x5 2D phase encoding[6] was used to minimise T2* decay, improving spectral resolution. The GIRF was constructed using 100 ms readouts. Both the self and B0 cross-terms of the GIRF were considered[7]; linear cross-terms were not included in this work. Figure 1a shows the magnitude of the measured ‘x’ gradient response functions. All code relating to the GIRF sequence creation (via PyPulseq[8]), processing, and calculation is available at https://github.com/jbbacon/GIRF_PE_Python.
Field perturbations occurring during the SVS readout, arising from gradient-induced eddy currents were predicted (Figure 1b-d). This was achieved by:
1) Multiplying the frequency domain representation of the gradient waveforms by the GIRF to estimate the actual gradient fields during the sequence, including during the readout.
2) Time-integrating the estimated gradient fields during the readout to compute the accumulated phase error from system imperfections.
The correction was performed by subtracting the accumulated phase from the measured phase of the non-water suppressed spectrum. The correction process was performed retrospectively, offline, in Python. Figure 2 displays the spectrum measured at a 20 mm ‘x’-offset the phantom. The water peak signal remains ~1e4 times stronger than the metabolite signals, and the GIRF correction considerably reduces sidebands, yielding a spectrum comparable to that with water suppression. Similar results were found at isocenter and for offsets in the ‘y’ and ‘z’ directions.
Figure 3 displays the in vivo spectra at isocenter and in the primary motor cortex. Although the correction at isocenter performs better, sideband reduction is evident in both cases.
FSL_MRS[9] was used to fit the metabolite concentrations in the GIRF corrected and the water suppressed spectra from the primary motor cortex. The results are displayed in Figure 4. Outside of a larger baseline and an over prediction of the metabolite concentrations between 3.5-4 ppm, the GIRF corrected spectrum performs comparably to the water suppressed spectrum. Our proposed method substantially reduces the impact of eddy current artefacts in non-water suppressed SVS, resulting in metabolite spectra that are comparable in quality to those obtained using water suppression. The in vivo results at isocenter suggest that the correction using the B0 cross terms of the GIRF works particularly well, whilst the reduced performance in the primary motor cortex suggest the correction using linear terms needs further correction, such as consideration of the linear cross terms
This method requires no additional hardware, and is generalizable to any SVS pulse sequence. The GIRF is acquired in a one-time calibration measurement, independently of the spectra, and can be used to predict the accumulated phase errors for any sequence with known input gradients. This method may potentially be extended to achieve non-water suppressed MRSI. This study introduces a new post-processing method for achieving non-water suppressed SVS. The method uses the GIRF to correct artefactual sidebands on the water peak by characterising the system imperfections from which they originate. This method does not require additional hardware and is generalisable to any SVS sequence.
James B BACON (Oxford, United Kingdom), Peter JEZZARD, William T CLARKE
11:00 - 12:30
#47726 - PG474 CRYO-CEST: Non-invasive imaging of cryoprotectants using chemical exchange saturation transfer.
PG474 CRYO-CEST: Non-invasive imaging of cryoprotectants using chemical exchange saturation transfer.
Cryopreservation holds promise for transplantation medicine via improved utilization and immunological matching of organs [1]. Successful organ cryopreservation and transplantation has recently been achieved in rodents, using vitrification [2-5]. Vitrification enables ice-free cryopreservation by replacing high proportions of tissue water with polar solvents (cryoprotective agents, CPA) to avoid water crystallization at cryogenic temperatures [6]. CPA introduction into organs via perfusion of the vasculature has to be precisely controlled: The minimal CPA concentration needed to vitrify must be exceeded in all parts of the organs to preclude damage from ice-formation during cooling and rewarming. At the same time, CPA toxicity correlates with increasing CPA concentration, exposure temperature, and time [7]. Therefore, techniques for precisely determining the moment of sufficient, but not excessive cryoprotectant equilibration in all parts of an organ before cryopreservation for transplantation would be of great utility. Typically employed CPAs for vitrification include ethylene glycol (EG), dimethyl sulfoxide (DMSO), and formamide (FA) [8]. While non-invasive x-ray computed tomography imaging of DMSO is possible due to the radiodensity of its sulfur atom [9, 10], FA and EG are only composed of CHON elements.
All three CPAs were analyzed individually. For this purpose, different amounts of concentrations (2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22 %w/v) were combined in 15ml phantom tubes with distilled water. Image acquisition was performed on a 7T system, by using a 3D snapshot CEST sequence with three different B1 levels (0.4, 0.6, 0.9 muT). CEST data were acquired from -4 to 4 ppm with 0.1 ppm sampling. To correct for B₀ and B₁ field inhomogeneities, WASABI [11] was employed to simultaneously obtain B₀ and relative B₁ maps. In order to calculate the MTRasym, an equidistant sampling was used for the Z-spectrum acquisition. An omega-plot [12] was calculated to determine the different concentrations in each tube retrospectively according to 1/MTRasym vs. 1/ω12. Since the slope of each line in the omega plot is inversely proportional to the concentration of the exchanging proton pool, the concentration can be directly inferred from the slope. Figure 1 shows for FA and EG that the peaks in both the Z-spectrum and the MTRasym appear to correlate with the respective concentration. While FA has the advantage to be very sensitive with concentration changes, EG has the advantage to resonate near resonance frequency of water and thus have less contributors through the NOE, ranging from -1 to -5 ppm. However, in the case of DMSO, additional NOE appear to occur which interfere with an adequate evaluation by the MTRasym.
Figure 2 shows the relative concentration using FA as an example, calculated from the slope of the omega plots at 2.6 ppm. Based on the known concentration of phantom tube 11 (22 % w/v), all other values were calculated relative to this reference concentration. The in vitro experiment shows that CPAs can be detected by CEST imaging. Assuming that no further MT effects occur in the Z-spectrum, the relative concentration can be calculated from the MTRasym. It has been demonstrated that EG oscillates in close proximity to the resonance frequency of water. Consequently, it is less susceptible to interference from NOE factors. However, EG shows a slight drift of the amplitude in MTRasym. FA reacts even more sensitively to changes in concentration and could therefore be a promising CPA marker. Further studies on in vivo tissues such as porcine organs are planned to verify the effect by including further MT effects and performing CEST imaging before and after CPA loading. In conclusion, cryoprotectants can be detected using CEST imaging. This seems to be a promising approach to control the amount of CPA and to test it further, for example in animal models.
Jan SCHÜRE (Nürnberg, Germany), Moritz ZAISS, Arnd DÖRFLER, Alexander GERMAN
11:00 - 12:30
#45929 - PG475 Development and Validation of an Open-Source Pulseq-Based MRE Sequence Using Experimental and Finite Element Phantom Data.
PG475 Development and Validation of an Open-Source Pulseq-Based MRE Sequence Using Experimental and Finite Element Phantom Data.
Magnetic Resonance Elastography (MRE) is a noninvasive imaging technique used to assess the mechanical properties of soft tissues by visualizing the propagation of externally induced shear waves through the tissue [1]. MRE enables the estimation of viscoelastic parameters such as stiffness, which are valuable for diagnosing and monitoring a variety of pathological conditions, including liver fibrosis[2], tumors[3], and neurodegenerative diseases [4]. The reliability of MRE depends heavily on the accuracy of both the pulse sequence used to acquire motion-sensitive images and the inversion algorithms that convert the wave image into Stiffness maps [5]. To support the development and validation of novel MRE pulse sequences and inversion techniques, phantom studies offer a controlled environment with well-characterized mechanical properties. In this work, we developed an open-source MRE pulse sequence based on the Pulseq framework. The sequence was tested experimentally on a homogeneous MRE phantom over a frequency range of 50–120 Hz using a 3T MRI scanner. In parallel, a finite element (FE) model of the phantom was created in FEBio[6] to simulate the wave propagation under identical boundary conditions and excitation frequencies. This dual experimental and simulation approach enabled a comprehensive comparison of the measured and simulated displacement fields, providing a robust framework for validating the custom Pulseq-based sequence.
A homogeneous cylindrical phantom (diameter 150 mm, height 130 mm; CIRS Inc., VA, USA) was imaged on a 3T MRI scanner using a custom MRE sequence developed with the Pulseq within MATLAB. The sequence employed a gradient-echo echo-planar imaging (GE-EPI) readout and incorporated motion-encoding gradients (MEGs) synchronized with externally applied mechanical vibrations. MEGs were applied in the through-plane (Z) direction with an amplitude of 10 mT/m. Mechanical excitation was delivered using a pneumatic active driver system (Resoundant Inc., USA) connected to a passive driver placed on the phantom’s top surface (Fig. 1). The phantom was positioned within a 32-channel head coil, and sinusoidal shear waves at 50, 60, 70, and 120 Hz were induced and synchronized with the MRE acquisition. For all frequencies, the driver amplitude was fixed at 30%. Image acquisition was performed using a single coronal slice positioned at the isocenter of the phantom. Displacement fields were encoded over a single vibration cycle to capture transient shear wave propagation, and motion-induced phase shifts were reconstructed into phase-difference maps. For finite element (FE) simulation, the phantom geometry was meshed using the GIBBON toolbox within MATLAB and imported into FEBio (Fig. 2). The phantom was modeled with a Young’s modulus of 9 kPa, Poisson’s ratio of 0.49, and density of 1000 kg/m³. To match the experimental boundary conditions, nodes on the lateral cylindrical surface were constrained in the X and Y directions, while a single-cycle sinusoidal displacement (amplitude 150 µm) was applied in the Z direction at the same four frequencies. Transient time-domain simulations were conducted. Phase images from the experimental and simulated datasets were compared (Fig. 3) at selected time points for each frequency (50, 60, 70, and 120 Hz). Overall, there was good qualitative agreement in wavefront shape and propagation direction. At 50–70 Hz, both datasets showed coherent wave patterns with similar wavelengths. At 120 Hz, increased attenuation and shorter wavelengths were observed, with the experimental data showing more noise and edge artifacts. These results indicate reasonable alignment, though some differences were evident, particularly at higher frequencies. The agreement between experimental and simulated phase images indicates that the Pulseq-based MRE sequence and FE simulation can capture key features of shear wave propagation in the phantom. Similar wave behavior was observed across all frequencies; however, discrepancies in amplitude and edge effects, especially at higher frequencies, suggest limitations in the current setup. These may result from unmodeled factors such as imperfect driver-phantom coupling, material property mismatches, or oversimplified boundary conditions. Overall, the results are promising but highlight the need for further refinement of both the sequence and simulation for improved accuracy. This study demonstrates the feasibility of using an open-source Pulseq-based MRE sequence in combination with finite element simulations for phantom validation. While qualitative agreement in shear wave propagation was observed across frequencies, discrepancies in wave amplitude and boundary effects indicate that further refinement of both the sequence and simulation setup is needed. This framework provides a solid starting point, but additional work is required to improve accuracy and robustness for broader application.
Mehmet Nebi YILDIRIM (Cardiff, United Kingdom), Samuel EVANS, Daniel GALLICHAN
11:00 - 12:30
#46902 - PG476 3D golden-means PROPELLER: Towards high spatio-temporal volumetric MRI.
PG476 3D golden-means PROPELLER: Towards high spatio-temporal volumetric MRI.
PROPELLER MRI [1] is a prominent MRI acquisition technique, which samples k-space in rotating blades. Because of the repetitive acquisition of k-space center, this method enables estimation of translational and rotational in-plane motion from the oversampled region in k-space. Moreover, combined with a golden-angle sampling scheme, this leads to a high spatio-temporal resolution because of the uniform angular distribution of PROPELLER blades.
There exist 3D implementations of the PROPELLER MRI scheme, e.g., 3D GRASE PROPELLER [2], which acquires a stack of PROPELLER blades (so called bricks) along the partition direction. The major drawback of these methods is that oversampling of k-space center is still restricted to the in-plane direction. Particularly, these techniques do not allow an extension of the in-plane motion correction scheme to through-plane motion correction and suffer from a low spatial resolution along the partition direction for large imaging volumes.
In this work, we verify the feasibility of a 3D golden-means PROPELLER acquisition scheme, which naturally extends the original 2D PROPELLER to three dimensions and enables high-spatio temporal resolution in all directions. Particularly, we extend golden-angle 3D GRASE PROPELLER to rotate about all three physical axes and apply our method to in-vivo cranial MRI.
Following [3], we generalize the 3D golden-means radial scheme to a 3D golden-means PROPELLER sampling scheme, i.e., the azimuth and polar brick angles are derived from the eigenvalues of the Fibonacci matrix (see Figure 1). The pulse sequence is adapted from a custom 3D GRASE PROPELLER sequence implemented in gammaSTAR [4].
We generalize the phase correction scheme in [1] to compensate for non-uniform brick centers arising from eddy currents and defective gradients in all three dimensions. To this end, we extend the originally defined 2D rhomboid-shaped low-pass filter to a 3D rhomboid-shaped filter, which suppresses low-frequency phase variations in image space, i.e., centering the DC component of each brick.
For reconstruction, we apply direct Fourier inversion using density compensation. Here, the forward operator is defined as the non-uniform discrete Fourier transform, which encodes the transformation from a uniformly resolved 3D imaging volume to the corresponding 3D golden-means PROPELLER k-space. Individual coil images are combined using the sum-of-squares approach.
Our experiments were performed on a healthy subject (male, 27 years old) using a Siemens Magnetom Vida Fit 3T system. We chose 4 s TR and 26.9 ms TE with a brick size of 96x32x32 and an FOV of 256 mm x 256 mm x 256 mm. Slice thickness was chosen as 8 mm to achieve a uniform k-space sampling in all directions per brick. A total of 30 bricks were acquired within 2 minutes total acquisition time and the final image matrix has size 96x96x96. Figures 2, 3, and 4 depict different reconstructed imaging slices along the sagittal, coronal and axial plane respectively. Our results suggest that 3D golden-means PROPELLER is a feasible 3D acquisition method for high spatio-temporal imaging in cranial MRI (see Figures 2-4). The straightforward extension of the phase correction to 3D is a promising result for future extensions of other PROPELLER-related methods. In future work, specifically the PROPELLER in-plane motion correction scheme must be extended to allow motion correction along all physical axes in 3D golden-means PROPELLER to enable robust distortion-free imaging.
3D golden-means PROPELLER enables comparatively high-resolution imaging in all dimensions for large imaging volumes compared to the standard 3D GRASE PROPELLER, because in the latter the repeated refocusing leads to low signal in outer partitions.
Our reconstruction algorithm is basic in the sense that the forward operator does not incorporate imperfect measurement conditions. A 3D off-resonance correction scheme must be developed to compensate for extra phase accumulation along the phase encoding directions to improve image quality. Furthermore, the oversampling of k-space center can further be exploited to severely reduce acquisition time by applying parallel imaging techniques.
Lastly, our method is a promising candidate for SNR- and motion-sensitive applications such as Arterial Spin Labelling, since it can acquire a large imaging volume within a low number of shots and should be tested on other organs of interest which move substantially during acquisition, e.g., liver. In summary, our results verify the feasibility of 3D golden-means PROPELLER in cranial MRI. It is a promising candidate for motion- and SNR-sensitive tasks which require high spatio-temporal resolution.
Tom LÜTJEN (Bremen, Germany), Jörn HUBER, Matthias GÜNTHER, Daniel HOINKISS
11:00 - 12:30
#46944 - PG477 Prospective undersampling compensation in motion-resolved abdominal PROPELLER MRI.
PG477 Prospective undersampling compensation in motion-resolved abdominal PROPELLER MRI.
Axial MRI of abdominal organs under free-breathing is a highly desirable imaging technique for patients who cannot suspend respiration. However, the patient’s respiration leads to severe motion during acquisition. A prominent motion-robust MRI acquisition technique is PROPELLER [1], which samples k-space in rotating blades. Particularly, oversampling of k-space center allows to estimate in-plane rotational and translational motion for each blade [1].
However, in axial imaging, respiration primarily induces through-plane motion, which cannot be corrected. To compensate for through-plane motion, one can resolve imaging data in an extra motion dimension, i.e., partition blades into different respiratory phases and reconstruct an image for each state separately. But this partitioning, especially considering the scarcity of shots in PROPELLER MRI, leads to severe undersampling per motion state, which cannot be sufficiently compensated for using already existing reconstruction frameworks, e.g., XD-GRASP [2].
In this work, we propose to adapt the rotation angle of each blade based on the current motion state to achieve a golden-angle sampling scheme per motion state, mitigating undersampling in motion-resolved PROPELLER MRI without relying on specialized retrospective reconstruction schemes.
The custom 3D GRASE PROPELLER [3] sequence and real-time feedback are implemented in gammaSTAR [4] and experiments were performed on a Siemens Magnetom Vida Fit 3T system.
Before each blade excitation, a global fat FID navigator is used to identify the current respiratory phase. Particularly, we use the center of mass (COM) of the FID signal along the coil dimension as a surrogate for the current respiratory phase.
Based on the current respiratory phase and the desired motion-resolution, we prospectively sort the next blade into the appropriate motion state. Specifically, we implement an online k-means algorithm with an experimentally determined threshold, which controls the maximal diameter of clusters (see Figure 1). A blade is sorted into a new motion state, if the desired motion-resolution is not achieved yet and the COM of its' corresponding FID lies outside of the maximal diameter of all existing motion states. Otherwise, the blade is sorted into the motion state, whose mean COM is closest to the COM of its' corresponding FID.
After clustering, the next blade angle is adapted during sequence runtime to the next golden-angle with respect to the current motion state. The resulting clustered PROPELLER data is reconstructed by means of direct Fourier inversion using gridding [1] and the least squares method, i.e., XD-GRASP without regularization.
Our method was verified in-vivo on a healthy subject (male, 25 years old) with a TR of 4 s and TE of 24.5 ms. The blade matrix size was 96 x 32 and 15 blades were acquired in total. Figure 1 shows the COM of FID navigators and the corresponding clustering.
Figure 2 depicts the COM of a train of FID navigators and the simultaneously acquired respiration signal detected by the systems’ Physiological Monitoring Unit (PMU).
Figure 3 shows the (non-) adapted PROPELLER trajectories.
Figure 4 shows different motion-resolved PROPELLER-based reconstructions. Our work verifies the effectiveness of the proposed prospective undersampling compensation in motion-resolved PROPELLER MRI.
The global fat FID navigator is a sufficient surrogate for true respiration, which does not require external hardware or a vendor-specific respiration tracking system (see Figure 2). However, in future work, the influence of preparation modules on the accuracy of the respiration estimation must be investigated.
The online k-means algorithm with thresholding yields a good separation between motion states and compensates for lack of reference data during early FID acquisitions but the threshold must be refined to sharpen the separation between motion states (see Figure 1).
Even low motion resolution can lead to severe undersampling in motion-resolved PROPELLER MRI, but our prospective blade angle adaptation effectively compensates for poor blade angle distribution (see Figure 3). Acquisition time can be reduced in the future by stopping the sequence automatically, when sufficient k-space coverage is achieved.
Prospective undersampling compensation allows us to reconstruct motion-resolved PROPELLER data without severe undersampling artifacts (see Figure 4). However, images still suffer from residual distortions arising from imperfect binning. Our method must be tested with higher motion resolution to minimize intra-bin motion, which can be further reduced by means of the PROPELLER in-plane motion correction.
Lastly, our method must be verified in motion- and SNR-sensitive applications such as free-breathing liver Arterial Spin Labelling perfusion MRI. In summary, our proposed method compensates undersampling in motion-resolved PROPELLER MRI effectively and allows a fair spatio-temporal resolution in free-breathing abdominal MRI.
Tom LÜTJEN (Bremen, Germany), Jörn HUBER, Matthias GÜNTHER, Daniel HOINKISS
11:00 - 12:30
#47718 - PG478 Comparison between diffusion-tensor and diffusion-weighted images along perivascular spaces index and influence of ROI size.
PG478 Comparison between diffusion-tensor and diffusion-weighted images along perivascular spaces index and influence of ROI size.
The glymphatic system (GS)[1,2] hypothesis aims to explain the waste clearance mechanism in the brain, which could be noninvasively evaluated using diffusion-tensor imaging (DTI) along perivascular spaces (ALPS) index[3]. A low index may suggest impaired GS function, as observed in neurodegenerative diseases[4].
Diffusion-weighted imaging (DWI) ALPS index could also be calculated when planning the imaging plane following the anterior commissure to posterior commissure line[5]. DWI is easier, faster, and more widely used in clinical routine than DTI, and it has shown its potential in the indirect evaluation of the GS in healthy volunteers[5] and for whole-brain radiotherapy[6]. Here, we assess how variations in the size of the region of interest (ROI) affect ALPS index calculation in DTI and DWI ALPS index.
Thirteen healthy volunteers (8 women; aged 22.8 ± 1.4 years) provided written consent before the examination. The study was approved by the local ethical commission.
We used a 3T MRI scanner (MAGNETOM Vida, Siemens Healthineers, Germany), with the following parameters for DTI and DWI: TE/TR = 92/4400 ms; FOV = 200 x 200 mm^2; voxel size = 2 x 2 x 2 mm^3; and b-values = 0 and 1000 s/mm^2. For DTI, 64 directions were used, and for DWI three orthogonal directions in the readout, phase-encoding, and slice-selection direction, with a total duration of 5:30 and 1:30 min for DTI and DWI, respectively.
The DTI ALPS index is calculated by using a ratio between diagonal elements of the diffusion tensor matrix (Dxx, Dyy, and Dzz) in two ROIs on the projection (proj) and association (assoc) fibers in the area of the periventricular veins as: mean(DxxProj,DxxAssoc)/mean(DyyProj,DzzAssoc)[3]. For DWI, the diffusivities along the x-, y-, and z axes were obtained directly from the scan and the ALPS index was calculated using the same formula[5].
The DTI and DWI were analyzed using DSI Studio[7] and ImageJ[8], respectively. For DTI ALPS, the ROIs (single voxel and 4-voxel isotropic) were positioned on a single slice according to a color-coded fractional anisotropy map by referring to susceptibility-weighted images. For DWI ALPS, In ImageJ, an image composite was generated with three channels: red, green, and blue for diffusivity along x-, y- and z- directions[5].
Statistical analysis was performed using Matlab R2022a (The MathWorks, Natick, MA) with α = 0.05. We investigated interhemispheric differences in the DTI and DWI ALPS indices and between 2 ROIs (Wilcoxon signed-rank). Intraclass correlation coefficient (ICC) was used to study the inter-method agreement and Bland-Altman plots to analyze the differences between methods and ROI configurations. No interhemispheric differences in DTI and DWI ALPS were found for 1- and 4-voxel ROI (p >> 0.05). Additionally, we did not observe any significant differences between DTI and DWI for both ROI sizes (p >> 0.05). However, inter-method agreement for 1-voxel ROI was poor (ICC = 0.3). The 4-voxel ROI showed moderate agreement between methods (ICC = 0.52). Bland-Altman plots (Fig. 2) suggest that while there is low bias between techniques, some variability does exist. We did not find significant interhemispheric differences in DTI and DWI and ROIs in healthy volunteers. However, larger ROIs should be preferred as they can cancel out the spatial inhomogeneities in the brain parenchyma[5,9].
Contrary to Taoka et al.[5], we did not find significant inter-method differences, and only poor-to-moderate inter-method reliability. The fact that we did not average the two hemispheric indices, which provided a higher ICC than a unilateral calculation[5], might explain this finding. The interhemispheric ALPS index average could be appropriate for research performed on healthy volunteers. However, several studies[10–12] have reported interhemispheric differences in the ALPS index. Therefore, when the area of the periventricular veins is intact, it is better to analyze the two hemispheres separately and to report possible asymmetries.
Additionally, inter-method variability should be considered in clinical or research settings to ensure consistent and reliable measurements Our findings highlight the importance of standardizing ROI configurations, thereby improving the reliability of DTI and DWI measurements.
If researchers need to compare their findings with other published results, then it is better to use a 4-voxel ROI, which shows the best agreement between methods.
Although recent developments have called the validity of the ALPS index into question, indicating that it should not be directly related to GS function, it might still represent a potential biomarker for diffusion in the perivascular space[13]. Indeed, various methods should be used in combination to study GS function.
Janina KREMER (Lübeck, Germany), Justus Christian RUDOLF, Aileen SCHMIDT, Peter SCHRAMM, Patricia ULLOA
11:00 - 12:30
#47832 - PG479 Comparison of deep learning predicted and universal pTx pulses for a 3D turbo spin echo sequence at 7T.
PG479 Comparison of deep learning predicted and universal pTx pulses for a 3D turbo spin echo sequence at 7T.
To counteract the problem of B1+ inhomogeneities arising at higher magnetic field strengths such as 7T, the concept of parallel transmission (pTx) was introduced. However, pTx pulses come with increased SAR exposure and require time-consuming calculation. To address the latter, pre-computed universal pTx pulses (UPs) [1] have been proposed to improve the excitation homogeneity in the human head without requiring online calibration. In order to enable tailored pTx pulse design in clinical routines at 7T, optimization times must be further reduced. Recently, deep learning (DL) approaches have been shown to be feasible for 2D single-slice excitations and either dynamic one-channel RF pulse design [2] or static pTx [3], providing negligible calculation time. To extend the potential of DL to multi-channel, dynamic pulse design, this study aims to investigate the use of a supervised trained neural network (NN) for scalable 3D dynamic pTx pulses for a turbo spin echo sequence. The resulting predicted pTx pulses are compared to UPs, representing the current state of the art without requiring additional calibration time.
A convolutional NN was trained using pre-acquired B1+ and B0 maps of the human brain to predict optimized RF and gradient waveforms. A total of 132 B1+ and B0 maps [4] were acquired with an 8Tx/32Rx head coil (Nova Medical, Wilmington, USA) at 7T (Magnetom Terra, Siemens Healthineers AG, Erlangen, Germany) and augmented retrospectively using affine transformations and elastic deformations [5], resulting in a total input library size of 5500. Corresponding target pulses, pulsestarget (Tpulse = 1.2ms, α = 10°), were optimized using a vendor-provided MATLAB toolbox with an interior-point based optimization algorithm [6]. The DL workflow is schematically illustrated in Figure 1. The NN was trained to minimize the mean squared error (MSE) between the predicted pulses ("pulsesDL ") and the corresponding pulsestarget. Subsequently, the model was used to predict pulses for 72 unseen test datasets. For comparison, two UPs (UPlowSAR designed to initialize individual pulse calculation and UPhighSAR designed for direct application without further calibration) were evaluated on the same test datasets. All pulses were designed using symmetric RF and anti-symmetric gradient shapes for scalability. For validation, an in vivo measurement of an unseen subject was performed. 3D T2-weighted SPACE images were acquired with the circularly polarized (CP) mode, DL predicted pulse and both UPs, each applied as both excitation and refocusing pulse (Δx3=0.4×0.4×0.5mm3, TE=299ms, TR=8000ms, TA=5:54min). Figure 2 compares the performance of the DL predicted pulses with the CP mode and the UPs evaluated for the 72 unseen data sets. The boxplots illustrate that the CP shim yields the highest median coefficient of variation (CV) with 25.9%. UPlowSAR reduces the median CV to 15.0%, while the pulsesDL further improve it to 14.1%. UPhighSAR achieves the lowest median CV of 12.1%. However, this comes at the cost of a higher specific energy dose (SED) of 9.1 mJ/kg, which is over 40% more than the median SED value of the pulsesDL (6.4 mJ/kg). UPlowSAR yields a SED value of 4.9 mJ/kg. Figure 3 shows the 3D T2-weighted SPACE images of a DL-wise “unseen” volunteer to validate the performance of the aforementioned pulses. While the CP mode and UPlowSAR fail to produce a uniform signal distribution across the whole brain, the pulsesDL and UPhighSAR accomplish a homogenous excitation without visible signal voids. While UPhighSAR slightly outperforms the DL-predicted pulses with a reduced median CV, it requires substantially more RF energy. In contrast, using an UP with reduced SED shows unacceptable performance compared to the pulsesDL in both simulation and measurement. With an average prediction time of a few milliseconds, DL-predicted pulses offer a fast solution for subject-specific pTx pulse design at 7T and can be beneficial in cases with anatomical anomalies. Nonetheless, there is room for improvement in terms of excitation uniformity. One reason for that might be insufficient generalization of the NN. Therefore, future work will focus on expanding the training dataset library and the implementation of a physics-informed loss function based on Bloch simulations [7]. In addition, combining the DL approach with a regular optimization for enhanced performance as in the FOCUS method [4] is conceivable, too. This study demonstrates the feasibility of using a DL approach for dynamic pTx pulse design, showing that DL-predicted pulses achieve comparable performance to state-of-the-art UPs. The results were verified both in simulation and in vivo measurement at 7T.
Sophia NAGELSTRAßER (Erlangen, Germany), Jürgen HERRLER, Mads Sloth VINDING, Nico EGGER, Patrick LIEBIG, Michael UDER, Moritz ZAISS, Armin NAGEL
11:00 - 12:30
#47608 - PG480 MRI-guided laser interstitial thermal therapy thalamotomy for patients with pharmacoresistant tremor: a single-center MR Imaging and MR Spectroscopy follow-up.
PG480 MRI-guided laser interstitial thermal therapy thalamotomy for patients with pharmacoresistant tremor: a single-center MR Imaging and MR Spectroscopy follow-up.
Pharmacoresistant tremors can significantly impair functional abilities. Thalamotomy using Laser Interstitial Thermal Therapy (LITT), guided and monitored by Magnetic Resonance Imaging (MRI), is among the recent therapeutic alternatives designed to enhance the quality of life for these patients [1-2]. Although practiced in the USA and Europe since 2019, there are currently no published studies examining spectroscopic and metabolic changes following LITT thalamotomy. Therefore, the primary objective of this study was to combine MRI and proton Magnetic Resonance Spectroscopy (MRS) to monitor morphological, spectroscopic, and metabolic characteristics within the treated ventral intermediate nucleus (VIM) region of the thalamus following LITT thalamotomy.
A total of 29 patients treated with LITT at the University Hospital of Amiens (from March 2019 to date) were prospectively followed. Imaging and spectroscopic data were acquired preoperatively, immediately postoperatively, at postoperative days 2–7, at 6 months, 12 months, and beyond 12 months for some patients. MRI data included T1-weighted, T2 FLAIR, T2*, diffusion-weighted imaging, perfusion imaging, and 3D T1-weighted sequences. Proton single Voxel Magnetic Resonance Spectroscopy (MRS) was performed using a Point RESolved Spectroscopy (PRESS) sequence at three echo times (TE: 35 ms, 144 ms, and 288 ms). MRI analyses included volumetric quantification of T2-FLAIR hyperintensities and diffusion-weighted hyperintensities within the VIM region. MRS spectra were analyzed using LCModel software to quantify metabolite ratios, specifically Choline (Cho), N-Acetyl-Aspartate (NAA), Myo-Inositol (mI), Glutamine-Glutamate complex (Glx), lactate, and CH2 and CH3 phospholipids, normalized relative to Creatine (Cr). MRI analysis (Figure 1.C) revealed the presence of small hyperintense volumes immediately postoperatively on T2-FLAIR (mean: 0.104 ± 0.062 cm³) and diffusion-weighted sequences (mean: 0.225 ± 0.118 cm³). These hyperintensities increased in 100% of patients at postoperative days 2 to 7 on both T2-FLAIR (mean: 3.302 ± 1.712 cm³) and diffusion-weighted imaging (mean: 1.455 ± 0.806 cm³). Subsequently, these hyperintense regions markedly decreased by approximately 98% at the 6- to 12-month follow-up period on T2-FLAIR (mean at 6 months: 0.061 ± 0.042 cm³; mean at 12 months: 0.026 ± 0.029 cm³) and diffusion-weighted images (mean at 6 months: 0.020 ± 0.036 cm³; mean at 12 months: 0.016 ± 0.032 cm³).
MRS results demonstrated spectroscopic and metabolic changes based on metabolite ratio calculations [Creatine (Cr), N-acetyl-aspartate (NAA), Choline (Cho), Myo-inositol (mI), and Lactate (Lac)]. Specifically, the mI/Cr ratio was increased in 100% of patients immediately postoperatively and subsequently decreased by 12 months. The Lac/Cr ratio was elevated in 85% of patients either immediately postoperatively or at postoperative days 2–7, and although it subsequently decreased, a residual lactate signal persisted long-term in 71% of patients (Figure 1.D). The present results provide a comprehensive overview of the MRI-based morphological and spectroscopic-metabolic evolution within the VIM thalamic region treated by LITT. These findings demonstrate an initial marked fluctuation in imaging and metabolic biomarkers, followed by progressive stabilization over the long term after short-duration and low-intensity LITT thalamotomy. The combined MRI-MRS approach presented here appears more promising than conventional MRI alone for refining the procedural planning, monitoring, analysis, and clinical interpretation of this innovative minimally invasive neurosurgical technique. To our knowledge, this study is the first to offer detailed insights into the morphological and metabolic dynamics of the VIM region following LITT, providing crucial data for evaluating the efficacy and impact of this minimally invasive neurosurgical approach. Future studies, involving larger patient cohorts and longer-term follow-ups, are necessary to validate these findings. Additional research exploring the functional impact of LITT thalamotomy on distant motor regions will also be conducted.
Salem BOUSSIDA, David LAYANI, Mickael AUBIGNAT, Aurélien LAMBERT, Adrien PANERO, Romain DRAILY, Amandine OSAER, Simon BERNARD, Melissa TIR, Michel LEFRANC, Jean-Marc CONSTANS (AMIENS)
11:00 - 12:30
#47614 - PG481 MR Spectroscopy evidence of brain alterations in Long Covid patients with persistent neurological complications.
PG481 MR Spectroscopy evidence of brain alterations in Long Covid patients with persistent neurological complications.
A considerable number of COVID-19 patients exhibit persistent neurological symptoms, such as anosmia, ageusia, fatigue, impaired attention and concentration, language disturbances, and reduced working memory. However, the underlying pathogenesis remains incompletely understood, particularly concerning brain metabolic alterations associated with these persistent neurological complications. To investigate this, combined proton Magnetic Resonance Spectroscopy (MRS) and Magnetic Resonance Imaging (MRI) data were obtained from a cohort of patients with Long COVID and compared with healthy controls, focusing on identifying potential spectroscopic and metabolic abnormalities linked to COVID-19 persistent neurological disorders [1].
Proton Magnetic Resonance Spectroscopy (MRS) and Magnetic Resonance Imaging (MRI) data were acquired from 20 patients diagnosed with Long COVID and compared with 5 healthy control subjects. MRI acquisitions consisted of 3D T1-weighted spin-echo sequences with and without gadolinium contrast enhancement, gradient-echo T2* (or SWI), diffusion-weighted imaging (DWI), T2-Fluid Attenuated Inversion Recovery (T2-FLAIR), coronal T2-weighted imaging, and Time-of-Flight Magnetic Resonance Angiography (TOF-MRA). Single voxel MRS was performed using the Point RESolved Spectroscopy (PRESS) sequence with three different echo times (TE: 35 ms, 144 ms, and 288 ms). Spectroscopic measurements were performed on three brain regions: medial frontal cortex, hippocampus, and pons. MRS spectra were analyzed using LCModel software to quantify metabolite ratios, including Choline (Cho), N-Acetyl-Aspartate (NAA), Myo-Inositol (mI), Glutamine-Glutamate complex (Glx), lactate, and CH2 and CH3 phospholipids, relative to Creatine (Cr). This study demonstrates the potential of proton MRS to detect spectroscopic and metabolic brain abnormalities in patients experiencing persistent neurological symptoms following COVID-19 infection. Compared to conventional MRI, MRS exhibited greater sensitivity in identifying and delineating COVID-19-associated cerebral alterations. Significant spectroscopic abnormalities were observed in Long COVID patients compared to healthy controls, varying according to brain region. These abnormalities suggest the involvement of multiple pathophysiological processes, including: (1) neuroinflammation (Figure 1), (2) glutamatergic dysfunction, (3) energy metabolism dysfunction, and (4) early neuronal dysfunction. The findings of this study provide evidence of spectroscopic, tissue-specific, and metabolic brain abnormalities that are measurable by Magnetic Resonance Spectroscopy (MRS) but not detectable on conventional MRI alone. These abnormalities indicate that SARS-CoV-2 infection can cause persistent metabolic alterations in specific brain regions, lasting several months after the acute phase of COVID-19. These data enhance our understanding of the complex pathophysiological mechanisms underlying persistent neurological symptoms observed in patients with Long COVID [2]. This study demonstrates that SARS-CoV-2 infection is associated with long-term cerebral metabolic disturbances that persist beyond the acute COVID-19 phase and are detectable and quantifiable by MRS, though undetectable by conventional MRI. These findings emphasize the clinical relevance and potential utility of incorporating MRS into routine clinical assessments and follow-up care for COVID-19 patients presenting persistent neurological symptoms.
Salem BOUSSIDA, Antoine GALMICHE, Yousef EL SAMAD, Amine ZEMANI, Nicolas DELEVAL, Julien MAIZEL, Serge METANBOU, Daniela ANDRIUTA, Olivier GODEFROY, Valery SALLE, Ahed ZEDAN, Catherine LE BRAS, Claire ANDREJAK, Jean-Marc CONSTANS (AMIENS)
11:00 - 12:30
#47846 - PG482 Fat/Water Spectroscopic Imaging for Improved Detection of Cribriform Prostate Cancer.
PG482 Fat/Water Spectroscopic Imaging for Improved Detection of Cribriform Prostate Cancer.
Cribriform prostate cancer is an aggressive subtype associated with higher risks of tumour metastasis, recurrence post-treatment and disease-specific mortality[1]. Identifying these highly heterogeneous tumours with sufficient sensitivity is challenging using established clinical diagnostic methods such as multiparametric MRI-targeted biopsy[2,3]. Fatty acid biosynthesis has been proposed as an important metabolic pathway for distinguishing cribriform lesions from other subtypes. However, invasive measurement of these fatty acids is impractical as tumours are typically small and difficult to identify using MRI. We assessed the potential for 2D 1H magnetic resonance spectroscopic imaging (1H-MRSI) to non-invasively differentiate cribriform from non-cribriform tumours in vivo. It is hoped that earlier and more reliable detection of cribriform prostate cancer may aid in identifying appropriate treatment and improve clinical outcomes for patients[4,5].
12 patients underwent prostate MRI (3 T Premier, GE HealthCare, Waukesha WI) using the in-built 60-channel posterior spine coil and a 30-channel anterior flexible array coil (GE HealthCare). Written informed consent was obtained from all patients as part of a prospective study approved by NRES East of England, Cambridge South (16/EE/0346).
Tumour localisation was achieved with a high resolution axial T2-weighted sequence over the prostate (FOV 18 cm) and diffusion-weighted imaging (FOV 28 cm, b = 1400/750/100 s/mm^2). Fat/water content was assessed with 2D axial MRSI through the tumour (Hamming-filtered density weighted trajectory over 3231 points of k-space, FOV 32 cm, matrix 64x64 interpolated to 128x128, 30° flip, TR 88.7 ms, TE 2.1 ms, 5 kHz full-receiver bandwidth). Images were co-registered to the high-resolution axial T2 series.
The fat fraction of the total signal was calculated from MRSI by integrating baseline-corrected magnitude spectra using Matlab R2024b (Mathworks, Natick MA) between 3.0 – 6.1 ppm (water) and -0.2 – 2.9 ppm (fat). ROIs were drawn on axial T2-weighted images by a radiologist with 13 years of experience in clinical prostate MRI reporting. Statistical significance was assessed by comparing: 1) the ratio of tissue ADC values to that of urine and 2) fat fraction measurements between cribriform lesions, non-cribriform lesions and contralateral healthy prostate tissue using a Mann-Whitney U test. Single axial slices from T2- and diffusion-weighted imaging series in a single patient are shown in Figure 1A-B, highlighting a left-sided peripheral zone tumour at the mid-point of the prostate. Lesions presented with low signal on T2-weighted images and high signal in diffusion-weighted images, both due to higher cellularity. Example images showing fat fraction of total signal from 2D MRSI within the prostate are shown in Figure 1C-D, with regions of adipose, muscle, healthy prostate and tumour highlighted for reference. Corresponding fat and water spectra from these regions are shown in Figure 1E-H.
The relative ADC value from diffusion-weighted imaging and the mean fat fraction as determined from 2D MRSI for cribriform and non-cribriform lesions and healthy prostate tissue across the patient cohort are shown in Figure 2. Fat and water peaks were clearly distinguishable in 2D MRSI magnitude spectra, with an elevated fat signal observed in cribriform lesions relative to non-cribriform disease. Reliable characterisation of cribriform lesions from other tissue types was possible (p < 0.05). No statistically significant difference was observed between non-cribriform lesions and healthy tissue using MRSI, and diffusion-weighted imaging was unable to reliably distinguish cribriform and non-cribriform lesions. These results broadly agree with a recent study correlating prostate cancer risk with fat fraction derived from a multi-echo imaging method[6].
The high spatial resolution of 2D MRSI employed in this work necessitated a comparatively long scan duration of 4:47 min for a single slice. However, the combination of high-resolution T2- and diffusion-weighted imaging facilitated reliable tumour slice identification, reducing the need to cover the entire prostate prospectively using MRSI. An increase in scan duration of approximately 5 minutes was considered clinically tolerable, suggesting that the incorporation of fat/water spectroscopic imaging into clinical protocol for active surveillance is feasible. Quantitative comparison of 2D MRSI with established fat/water imaging methods in the prostate in future patients may be of interest. We have demonstrated the feasibility of using elevated fat fraction as a non-invasive biomarker for cribriform prostate cancer with MRSI. Multi-centre validation of this approach may facilitate integration into routine clinical practice to improve the reliability of cribriform prostate cancer detection and associated clinical outcomes.
Jonathan BIRCHALL (Cambridge, United Kingdom), Nikita SUSHENTSEV, Mary MCLEAN, Anne WARREN, Tristan BARRETT, Ferdia GALLAGHER
11:00 - 12:30
#45947 - PG483 Normalization-free Quantitative Analysis of Xe-129 HyperCEST Image Series.
PG483 Normalization-free Quantitative Analysis of Xe-129 HyperCEST Image Series.
The combination of hyperpolarized Xe-129 and chemical exchange saturation transfer as HyperCEST MRI offers unprecedented sensitivity to detect molecular targets with xenon biosensors at nanomolar concentrations.[1] Despite the 10000-fold higher spin polarization of Xe-129 after spin exchange optical pumping (SEOP), the signal-to-noise ratio (SNR) of dissolved Xe is still low compared to proton MRI of aqueous samples because of a low spin density ([Xe-129] < 100 µM) in most experimental settings. Moreover, the non-self-renewing character of hyperpolarized magnetization requires re-delivery of fresh Xe for each data point in a z-spectrum that is derived from HyperCEST MR image series. This imposes challenges because the quantitative evaluation usually requires a defined and stable baseline for normalization. SEOP setups may deliver a variable starting magnetization from shot to shot, particularly during warm-up, that is not immediately seen in the MRI scans. Here, we present a method that circumvents normalization and delivers reliable quantitative information even for rather unstable baselines.
Experiments were performed at 9.4 T on a Bruker Avance III HD console, using a gas mixture of 5% natural abundance Xenon, 10% N2 and 85% He dispersed into a concentric two-compartment phantom at an operating pressure of 4.5 bar (abs.). MDA-MB-231 cells were labelled with cryptophane cages that were functionalized with a peptide sequence for addressing the LHRH receptor and served as targeted HyperCEST agent for reversibly bound Xe. The phantom’s inner compartment (IC) contained 300 µL with 500'000 labelled cells/mL, the outer compartment (OC) served as a control with 500 µL of unlabeled cell suspension (same cell density). The Xe gas mixture exists the SEOP setup with a continuous flow of 300 mL/min and was dispersed into the phantom for 15 s prior to a 5 s wait period and subsequent HyperCEST acquisition with a RARE sequence. The saturation offset was incremented stepwise to cover the range of the expected HyperCEST response. Signals for z-spectra were obtained by evaluating ROIs with the scanner's image sequence analysis tool. The signal baseline is particularly unstable at the beginning of SEOP operation (Fig. 1). However, the reference signal from the OC can be approximated by a polynomial behavior that is fed into the fitting of the HyperCEST response in the IC with the full HyperCEST (FHC) model.[2,3] Fit results were compared to another z-spectrum that was taken later with a stable baseline (Fig. 2). The signal of ROIs from the two compartments exhibits a standard deviation (SD) of ca. 20%. However, changes outside the HyperCEST response are strongly correlated between both compartments. The OC reference signal can be approximated by a 3rd or 5th order polynomial (P3/P5). Using P5 with a scaling factor as the baseline for the IC signal yields fit results that are extremely consistent with a data set that exposes only a weak linear change in the baseline (Fig. 2). In particular, depolarization rates and line widths are in perfect agreement between these measurements and the HyperCEST intensity is highly consistent for different saturation times. The polynomial approximation of the baseline should be done carefully and may not be used outside the acquired data range. Nevertheless, P5 for the reference signal is a suitable input to approximate the baseline in the target volume and achieve a HyperCEST quantification with a standard deviation that is ca. 10-fold smaller than the SD of the ROI signals. The correlated signal change throughout the z-spectrum would thus allow to identify HyperCEST responses as small as ca. 2-3%. Distorted baselines are not necessarily a limitation for reliable quantification of HyperCEST data. Given the availability of a reference ROI, this approach should be valuable for future biomedical applications with low SNR where a reference signal can be used as input to compensate for variable baselines. It also allows to quantify less ideal datasets that were previously discarded due to inappropriate normalization.
Leif SCHRÖDER (Heidelberg, Germany), Jabadurai JAYAPAUL
11:00 - 12:30
#47721 - PG484 Fast Mapping of Intramyocellular Lipids Using Spiral MRSI and novel Apparent IntraMyocellular Lipids Content Indicator (AIMLI): Development, validation and Application for Longitudinal Metabolic Monitoring.
PG484 Fast Mapping of Intramyocellular Lipids Using Spiral MRSI and novel Apparent IntraMyocellular Lipids Content Indicator (AIMLI): Development, validation and Application for Longitudinal Metabolic Monitoring.
The quantification of intramyocellular lipids (IMCL) is of major interest in metabolic studies. Still, it remains constrained by long acquisition times, complex post-processing, and a lack of spatial resolution when using conventional spectroscopy [1], [2], [3]. This study introduces a rapid and straightforward method using proton spiral MRSI and a novel Apparent IntraMyocellular Lipid Indicator (AIMLI) to effectively map lipid distributions in muscle tissue. AIMLI was evaluated through numerical simulations accounting for susceptibility effects, validated in healthy volunteers against standard quantification techniques [4], and applied in a longitudinal study to monitor metabolic changes due to prolonged fasting.
Spiral MRSI data were acquired using a 3T clinical scanner (MAGNETOM Prisma, Siemens Healthineers) using a custom sequence with spatially selective excitation followed by a spiral readout gradient (FOV=200×200×25mm³, spatial resolution=64×64, voxel size=3.1×3.1×25mm³, TR/TE=2000/2ms, 1024 points, temporal resolution=500ms, spatial/temporal interleaving=22/5). Total acquisition time was 3min48s per slice. A 1H/31P transmit/receive surface coil (Rapid Biomedical) was positioned under the calf muscle of healthy volunteers.
Post-processing involved regridding to a Cartesian grid with 2× oversampling, Hamming apodization, and inverse 2D Fourier transform. Given the considerable spatial variations in phase and frequency, we put in place a robust spectral correction method, following the approach of Le Fur et al. [5], that utilized the strong water peak for phase alignment and frequency registration. The AIMLI was calculated from the cumulative amplitude curves of the modulus spectrum in the lipid region (1.1–1.7 ppm). Three chemical shift values (1.23, 1.31, 1.38 ppm) were used to generate maps reflecting apparent IMCL and EMCL contributions. These maps were used to assess repeatability, spatial distribution, and inter-subject variability. Full quantitative validation was performed in selected voxels using LCModel analysis [6] (figure 1).
We applied AIMLI for the monitoring of changes along a nutritional intervention [7] on 21 subjects, with MRSI scans acquired at baseline (D–1), post-fasting (D+12), and follow-up (D+30). Reference IMCL quantification was also performed using SVS-STEAM (TR/TE/TM = 3000/10/10 ms) and LCModel analysis [8]. Spatially averaged AIMLI values were extracted over muscle ROIs and compared across time points using non-parametric Friedman tests, with Wilcoxon signed-rank tests and Bonferroni correction for post-hoc analysis. Group-level AIMLI values demonstrated significant differences across the three time points (p<0.05). Post-hoc comparisons revealed a significant increase in mean AIMLI after fasting (D+12), followed by a return to baseline at D+30. These trends were consistent with those observed with SVS-LCModel data [8], supporting the physiological relevance of AIMLI (table 1 and figure 2). The AIMLI maps of two individual cases of special interest are presented in figure 3. In the first subject, AIMLI increased post-fasting and returned to baseline later, with spatial heterogeneity across different muscle heads (e.g., soleus, gastrocnemius). In a the second case with fat infiltration (incidental discovery in ours volunteers confirmed by Dixon imaging), AIMLI decreased progressively from D–1 to D+30. Notably, AIMLI revealed focal lipid changes in the medial soleus region that were not detected by fat fraction maps, likely due to Dixon’s sensitivity to EMCL over IMCL. Our findings demonstrate that AIMLI derived from spiral MRSI enables fast and reproducible mapping of apparent IMCL content with high spatial resolution. The modulus-based frequency registration ensures robust automated post-processing, reducing user intervention. Compared to SVS, AIMLI provides complementary spatial insights (modified ICML/(EMCL+IMCL) ratio of crucial interest at constant PDFF) and is more sensitive to subtle regional metabolic shifts. These advantages make it particularly suitable for longitudinal or interventional metabolic studies, and for detecting localized pathological alterations. The proposed ALCI method offers a fast, spatially resolved, and semi-quantitative alternative to traditional MRSI quantification approaches. Its clinical feasibility, low acquisition time, and resilience to spectral distortions position it a promising tool for metabolic imaging and follow-up in diverse physiological and pathological contexts.
Antoine NAËGEL, Antoine NAËGEL (Lille), Magalie VIALLON, Benjamin LEPORQ, Kevin MOULIN, Pierre CROISILLE, Hélène RATINEY
11:00 - 12:30
#47714 - PG485 Mapping of mobile macromolecules using metabolite-nulled sLASER with adiabatic water suppression and inversion and correlation with Macromolecular Proton Fraction: a 3T feasibility study.
PG485 Mapping of mobile macromolecules using metabolite-nulled sLASER with adiabatic water suppression and inversion and correlation with Macromolecular Proton Fraction: a 3T feasibility study.
Metabolite-nulled proton magnetic resonance spectroscopy (¹H MRS) reveals broad signals from mobile macromolecules (MM). While their exact molecular sources remain unknown, these signals show regional variation and are altered in pathology such as brain tumors (BT), and their growth disrupt the myelin structure [1]. Myelin breakdown products may become mobile and contribute to the MM spectrum. Another potential quantitative marker of myelin disruption could be the magnetization-transfer based macromolecular proton fraction (MPF) method [2], MPF shows a strong association with local myelin density in various neurological conditions [3]. Combining MM profiling from MRS with myelin mapping may provide complementary information, improve the characterization of BT-related damage, and allow for a better delineation of BT margins, which could improve treatment planning. This study is taking a step back from ultra-high-field MRSI applications for mapping brain MMs [4] and moving towards a potential clinical application at 3T. Its aim is to create a robust protocol to measure BT-associated demyelination by exploring correlations between MM signal intensities and myelin content quantified via MPF.
Data were acquired from two healthy volunteers using a 3T MRI (MR7700, Philips, The Netherlands) with a 16-channel head coil. The protocol included 3D T1w, 2D T2w and 3D FLAIR (12 min). For both single voxel (SV) and MRSI acquisitions, the sLASER sequence was applied, with an adiabatic inversion prepulse (hypsec, 750°, 7 ms, center at 2.5 ppm) introduced prior to VAPOR water suppression (WS). Due to low T1 of the MM, reducing TR improves the SNR of MM [3]. A minimum TR = 1.4 s was achieved by shortening WS delays and pulse durations by ~30%, yielding 145 Hz WS bandwidth and 520 ms WS duration. The sLASER parameters were TE = 31 ms, bandwidth = 2000 Hz, 1024 points. Assuming a metabolite T1 of 1.4 s [5], the inversion time (TI) was pre-estimated as 530 ms.
For the first volunteer, single-voxel MRS in the posterior cingular cortex (PCC) with VOI = 45×25×25 mm³, was acquired to determine the optimal TI, checking TI = 520, 530, and 540 ms (NSA = 192, 4.5 min each). Except for -Ch2 of Creatine, no metabolites were visible at 530 ms (Figure 1A). For the second volunteer, 2D MRSI was acquired at the PCC level using TI = 530 ms (NSA = 4, SENSE = 1.5×1.5, FOV = 240×240 mm², matrix size = 15x15, slice thickness = 20 mm, scan time = 12 min). The reconstructed voxel size was 7.5×7.5×20 mm³. MPF mapping [2] was acquired alongside MRSI with an EPI sequence (FOV = 230×230×182 mm³, voxel size = 0.7 mm³). MT settings: TR = 45 ms, TE = 4.6 ms, FA = 8°, MT offset = 1200 Hz, scan time = 3 min. Variable FA: TR = 20 ms, TE = 4.6 ms, FA = 20° and 4° (3 min). Surrogate B₁ mapping [6] was applied.
MRS(I) data were processed in jMRUI using AMARES, with Lorentzian peaks and prior knowledge adapted from [7]. MPF maps were generated using an open-access tool [8]. MPF values for each voxel were obtained with an in-house MATLAB code. MM amplitudes were then correlated with MPF values in the corresponding voxels. The metabolite-nulled SV spectrum from volunteer 1 is shown in Figure 1A. The AMARES fit demonstrates good fitting quality with low residual. Spectral fitting of an MRSI voxel from volunteer 2 is shown in Figure 1B. The MM and MPF maps obtained from Volunteer 2 are presented in Figure 2, illustrating spatial variations in MM signal intensities across the brain. Notably, regions with high MPF values (bright areas) correspond to low MM signal intensities (green and blue areas). Negative MM–MPF correlations were observed (examples in figure 3). In this pilot study, we demonstrate that the sLASER sequence, combined with both an adiabatic inversion prepulse and water suppression, is a method feasible for acquiring metabolite-nulled spectra and for mapping individual macromolecular (MM) components. At 3T, a spatial resolution suitable for tumor studies can be obtained within 12 min. The spatial patterns observed in our data, such as elevated MM signals in grey matter, are consistent with previous ultra-high-field findings. Although myelin is composed of lipids and specific proteins, these components are largely structurally bound and thus not highly mobile under physiological conditions [9]. The negative correlations between MM signal intensities and MPF values support this interpretation. The proposed acquisition protocol offers a unique opportunity to correlate the myelin breakdown measured by MPF with the increase in breakdown products measured by MM-MRSI in tumors. Further investigation will show whether the increased specificity will improve the diagnostic value of MPF in tumor tissue. Our findings support the feasibility of MM mapping using sLASER at 3T and open the opportunity to explore its role in tumor-related myelin disruption.
Andrei MANZHURTSEV (Frankfurt/Main, Germany), Nouha TEGHLET, Seyma ALCICEK, Dennis C. THOMAS, Ulrich PILATUS, Vasily L. YARNYKH, Elke HATTINGEN, Katharina J. WENGER
11:00 - 12:30
#47858 - PG486 Feasibility of the Detection of Human Hepatocellular Content of Lipids using Deuterium Metabolic Imaging.
PG486 Feasibility of the Detection of Human Hepatocellular Content of Lipids using Deuterium Metabolic Imaging.
To study hepatic disorders, assessment of hepatocellular content of lipids (HCL) using well-established single voxel ¹H-MRS [1] is of high interest. ²H(deuterium)-MR spectra contain analogous metabolic information but exhibit a relatively low natural abundance of ²H (0.012%), which potentially hampers the detection of HCL using deuterium metabolic imaging (DMI) [2]. Despite that, DMI offers good MR sensitivity due to short T1 relaxation times, and is less susceptible to magnetic field inhomogeneities [3]. One major advantage over ¹H-MRS is the ability to orally administer in low doses harmless ²H-labeled substrates, e.g. glucose or water to dynamically assess changes in ²H spectra, which could offer novel non-invasive insight into lipid metabolism during different interventions [4]. This study aimed at a DMI-based HCL estimation.
Four volunteers (sex: 1f/3m, age: 26-64y, BMI: 30.1±3.7kg/m²) with presumed high HCL were measured in a 7T MR system (Siemens Healthineers, Erlangen, DE) using a dual-tuned ¹H/²H surface coil (2 ²H-channels (~27x27cm), STARK CONTRAST MRI Coils Research, Erlangen, DE). The subjects were situated in a right lateral position with the liver centered on top of the RF coil.
Standard ¹H MRS-based HCL measurement was performed according to Gajdošík et al. [1] using a ¹H-GUSTEAU sequence at three different voxel positions (á 8 measurements, VOI: 3x3x3cm³) within the liver. Additionally, the ¹H methylene content (MC) was calculated analogously with solely its own resonance (1.3ppm) instead of all lipids.
For DMI an FID MRSI sequence with 3D density-weighted concentric ring trajectory readout (CRT) was used (2ms block pulse, 2ms acquisition delay, TR: 290ms, 47 circles, FOV: 27x27x26cm³, matrix: 22x22x21, total time: 8:21min) [5]. Anatomical images were acquired with a 3D gradient-echo sequence (scan time: 1:10min) for segmentation. 3D Slicer [6] was used to mask the liver and a smaller region within the hepatic tissue to calculate the ²H MC. Both volumes were down-sampled to the MRSI matrix size. The spectra from the liver voxels were further processed with an in-house programmed pipeline (MATLAB R2012b, LCModel v6.3, Python 3.12) for image reconstruction and spectral fitting. The used basis set for LCModel was generated previously, containing water (4.7ppm) and a single lipid methylene line (1.3ppm). Spectra from voxels within the smaller mask were averaged after phasing and frequency alignment using FSL-MRS [7] within the pipeline. Unlike for ¹H no T1 corrections were applied. The ²H MC was calculated as mentioned above and an extrapolated ²H HCL value was derived by applying the HCL/MC ratio from ¹H. Figure 1 shows the DMI-MRSI grid and the GUSTEAU-voxel location superimposed on anatomical images and depicts an averaged spectrum of the ¹H-MRS, the metabolic map of ²H-labeled water and (fitted) spectra of chosen voxels. The mask for spectral averaging of the DMI voxels and the averaged spectrum are depicted in Figure 2, including a comparison with the ¹H acquisition. With a nominal voxel size of 1.86ml, the accumulated volumes used to derive the lipid content ranged from 367 to 565cm³ (197–303 voxels). MC values of 6.23%/4.94%, 5.44%/3.81%, 4.01%/3.70%, and 9.45%/6.18% were calculated for ¹H/²H, respectively. These correspond to the (extrapolated) HCLs of 8.54%/6.77%, 7.45%/5.22%, 5.32%/4.91%, and 12.9%/8.40% (Fig. 3). At this stage, no significant difference (p=0.079), but high correlation (Pearson correlation: 0.961) was detected between the two methods. Low natural abundance of 2H limits the detectability of low HCL levels solely based on 2H signals [8]. The results show that the DMI-based assessment underestimated MC for all volunteers systematically by up to 35%. This deviation could result from the missing correction for incomplete relaxation, as there were no ²H T1 values of water and methylene available. Furthermore, errors could be introduced by the voxel selection process (masking), the pre-processing for averaging and by the fitting procedure (basis set), and partial volume errors. Therefore, DMI should not be used independently for static HCL assessment, but instead to assess complementary data on metabolic dynamics, e.g., after oral tracer administration and accompanied by other well-established 1H-based techniques [1], at best, in an multinuclear interleaved matter [9]. At this stage, the study is limited by the small number of subjects. The study demonstrates the feasibility of HCL assessment with DMI if a certain minimum lipid content is present in the liver. Exact level of sensitivity will be determined in further measurements. Nevertheless, offers the possibility to perform dynamic studies by 2H enrichment during different tracer intervention, e.g., administration with oral intake of heavy water to study hepatic de novo lipogenesis. Further optimization in hardware, acquisition and post-processing, could result in higher precision in relation to the ¹H HCL estimation.
Lorenz PFLEGER (Vienna, Austria), Viola BADER, Fabian NIESS, Bernhard STRASSER, Thomas SCHERER, Wolfgang BOGNER, Siegfried TRATTNIG, Peter WOLF, Martin KRSSAK
11:00 - 12:30
#47938 - PG487 New trajectories for functional MRSI.
PG487 New trajectories for functional MRSI.
Density weighted (DW) k-space sampling is often used in magnetic resonance spectroscopic imaging (MRSI) to reduce bleeding of the strong lipid signals present in the scalp into brain voxels. To achieve this, trajectories are typically designed for a point spread function (PSF) which minimises contributions from distant regions (minimise stopband ripple). This, in turn, can be achieved by having a sampling density close to the Hann window. DW concentric ring trajectories have proven effective for spectroscopic imaging [1, 2]. However, they are suboptimal for functional MRSI (fMRSI), as they require many repetition times (TRs) to form a single image, with sparse ring coverage leading to large gaps in k-t space. From fMRI BOLD imaging, we can assume any functional response diminishes over time as participants habituate to a given stimulus [3] (and references within). It is therefore desirable to implement trajectories that retain a Hann window density whilst comprehensively sampling k-t space in short time blocks.
To address these challenges, we propose two novel 2D DW k-space sampling patterns inspired by rosette and spiral imaging trajectories (Fig. 1) [4]. An additional motivation for this work is the emergence of non-water-suppressed MRSI techniques [5-7], where each TR could yield a B0 map to facilitate motion and shim correction [8]. Compared to conventional functional MRS (fMRS), fMRSI promises advantages including lower partial volume, improved spatial specificity, and reduced motion sensitivity.
Rosette trajectories were generated using the general formula used in [9], with the addition of a slowly varying rotation matrix. Spiral trajectories were constructed using the Spiral Gen support routine [10], and were extended to include analytical solutions for the time optimal return to the k-space centre. Three temporal interleaves were applied. Trajectories were rotated by the golden angle (137.5 degrees) between each TR.
Both trajectories were optimised via least squares regression, comparing their k-space densities with the Hann window. Density weighted concentric rings were also implemented for comparison. All trajectories were designed to reconstruct to a 32x32 image (FoV 240mm, in-plane voxel size 7.5mm, slice thickness 15mm).
Phantom (4 minutes, TR 1.5 s) and in-vivo (5 minutes TR 1.5s) studies were conducted at 7T (Siemens MAGNETOM 7T Plus, 8Tx32Rx Nova coil, 70mT/m, max slew 200 mT/m/ms). Identical 2π-CSAP magnetisation preparation (as implemented in [11]) was used. All sequences were generated using pypulseq [12]. For the in-vivo study, a slice which covers both motor and visual cortex was selected. These areas are commonly used in fMRS experiments [13]. Reconstruction was performed using BART [14] (coil sensitivity estimates and nufft adjoint with density correction [15] to Hann filter). The density of the optimised trajectories and their point spread functions are presented in Fig. 2, with the more flexible, variable density spiral achieving a closer match to the Hann window than the rosette. The concentric rings have the smoothest, and broadest central lobe. After density reweighting, each trajectory’s PSF is visible in its respective phantom reconstruction (Fig. 3B). The bright spot in each plot is unsuppressed water due to B0 distortion around a filling cap, acts as a high signal point source and so reflects the point spread function. Far from this water contaminant, in phantom the spectra obtained by all three trajectories was clean. Fig 3A shows the spectra from the phantom with normalised baseline noise standard deviation. Imaging rosettes and spirals approach the SNR of concentric rings despite the reweighting. Figure 4 shows results from in vivo acquisitions. Preprocessing and fitting of the in-vivo spectra are left as future work. In Figs. 3 & 4, density compensation is shown to smoothen and broaden the resulting image. All trajectories show similar PSF after this reweighting. Because spirals and rosette densities approach the desired Hann function weighting the loss of SNR is limited. While density compensation was shown reduces the lipid contamination toward the centre of the brain, it increases contamination at the edges, in the cortex where functional activation occurs (Fig 4.B). It therefore might be better to use un compensated or an inverse iterative reconstruction which will result in lower apparent SNR (due to smaller voxels), but less lipid bleeding. Future work and comparisons will determine the optimal route for fMRSI. Two novel trajectories for time-resolved MRSI are presented, a rosette and a variable density spiral. Both trajectory densities were optimised on the Hann window using L2 regression. Phantom and in-vivo studies were conducted to compare both trajectories to density-weighted concentric rings. This work lays the foundation of a sequence for (motion and shim) corrected time-resolved MRSI targeting functional metabolism.
Simon FINNEY (Oxford, United Kingdom), William CLARKE
11:00 - 12:30
#47940 - PG488 Role of peri-tumoural lipid composition for aggressive breast tumour using chemical shift-encoded imaging at ultra-high field of 9.4 T.
PG488 Role of peri-tumoural lipid composition for aggressive breast tumour using chemical shift-encoded imaging at ultra-high field of 9.4 T.
Breast cancer is the most common female cancer in the UK and globally, and the deregulation of lipid composition in the breast has been suggested as a risk factor [1,2]. Chemical shift-encoded imaging (CSEI) allows rapid lipid composition mapping, utilising the known resonant frequencies of lipids with a theoretical model for the quantification of monounsaturated, polyunsaturated and saturated fatty acids (MUFA, PUFA, SFA) [3,4]. However, the resolution is limited in clinical MRI due to a low signal to noise ratio (SNR) at a typical field strength of 1.5 – 3 T and the scan time tolerable by the patients. Ultra-high field preclinical MRI offers elevated SNR with typical ex vivo deployment, facilitating the acquisition of submillimetre lipid composition maps. We therefore hypothesise that deregulation in peri-tumoural lipid composition show a difference between tumour grade, an indicator for cellular differentiation and tumour aggressiveness, in breast tumour specimens.
We hence conducted high resolution imaging on specimens excised from patients with invasive ductal carcinoma using a 9.4 T preclinical MRI scanner. The study was approved by the North West – Greater Manchester East Research Ethics Committee (Identifier: 16/NW/0221), and signed written informed consents were obtained from all the participants (Figure 1).
Specimen Preparation: Twenty-one breast tumour tissue blocks, 12 Grade 2 and 9 Grade 3, were dissected into size of approximately 50 × 50 × 10 mm after routine histological examination. The tissue blocks were fixed in 10% formalin solution and stored in screwed-top plastic containers at 4˚C. Each tissue block was placed in a custom-made, standard size cassette holder layered with gauze and filled with a susceptibility-matched perfluorocarbon liquid (FluorinertTM, 3M, St. Paul, MN, USA), before the whole set up was sealed with a tightly fitted lid prior to MRI.
Lipid Composition Mapping: All images were acquired on a 9.4 T preclinical MRI scanner with a 20 cm diameter inner bore (Bruker, BioSpec, Ettlingen, Germany). The cassette holder set up was snug-fit inside the middle of a 12 cm quadrature coil (Rapid Biomedical, Rimpar, Germany), and inserted into the centre of the magnetic field. Lipid composition images were acquired using a 3D CSEI sequence [5,6] with 24 echoes, initial echo time of 2.14 ms, echo spacing of 1.03 ms, repetition time of 50 ms, reconstruction matrix of 256 × 256, and an isotropic resolution of 0.25 mm.
Image Analysis: Image analysis was conducted in MATLAB (R2023b, MathWorks Inc., Natick, MA, USA). A simplified triglyceride model was used to map the number of double bonds from the complex data on a pixel-by-pixel basis, and quantitative maps of MUFA, PUFA and SFA were subsequently derived as a fraction of the total amount of lipids [3,4]. The tumour boundary was delineated on the first echo of CSEI magnitude images, and the peri-tumoural region was defined as a 2 mm (8 voxels) annular ring surrounding the tumour boundary. The mean lipid composition from the region-of-interest was subsequently computed for each lipid constituent.
Statistical Analysis: All statistical analysis was performed in the R software (v4.3.2, R Foundation for Statistical Computing, Vienna, Austria). Wilcoxon rank sum tests were performed to compare the difference in lipid constituents between Grade 2 and 3 tumours. The correspondence between lipid constituents against tumour diameter was examined using Spearman’s rank correlation test. A p value < 0.05 was considered statistically significant. There was a significantly higher PUFA (p=0.028) of Grade 3 breast tumours (0.22, interquartile range (IQR): 0.21 – 0.22) in comparison to Grade 2 (0.20 (0.20 – 0.21)) (Figure 2b, Table 1). There was no significant difference in MUFA (p=0.095) between Grade 2 and Grade 3 (Figure 2a, Table 1). There was no significant difference in SFA (p=0.069) between Grade 2 and Grade 3 (Figure 2c, Table 1). There was no significant correlation in PUFA (rs = 0.09, p=0.710), MUFA (rs = 0.14, p=0.552) and SFA (rs = -0.14, p=0.542) against tumour diameter (Figure 3). Deregulation of peri-tumoural lipid composition was associated with tumour aggressiveness. PUFA is depleted in higher grade tumour [7], and may induce an increased concentration of PUFA in the peri-tumoural region to support elevated membrane synthesis during cancer progression [8]. However, an elevated mechanistic action in PUFA may not relate to morphological tumour size. There was an association between peri-tumoural PUFA and tumour grade. Lipid composition imaging at ultra-high field might have potential to probe cellular mechanistic actions in the tumour microenvironment.
Sai Man CHEUNG (Newcastle upon Tyne, United Kingdom), Kwok-Shing CHAN, Kangwa NKONDE, Yazan AYOUB, Nicholas SENN, Bernard SIOW, Jiabao HE
11:00 - 12:30
#46849 - PG489 Quantitative 7Li MRI in patients with bipolar disorder initiating lithium treatment: a first look at the European R-LiNK multicentric dataset using a region-based approach.
PG489 Quantitative 7Li MRI in patients with bipolar disorder initiating lithium treatment: a first look at the European R-LiNK multicentric dataset using a region-based approach.
Bipolar disorder (BD) is a debilitating psychiatric disease marked by extreme mood swings with a large societal burden. For decades, lithium salts have been prescribed to stabilize BD patients’ moods [1]. Although their effectiveness in preventing both manic and depressive phases has been demonstrated [2], the precise mechanism of action is still only partially understood. Moreover, only a third of BD patients fully respond to lithium treatment. It is in this context that the R-LiNK project [3-4] was initiated, gathering expertise across multiple domains including Psychiatry, Neuroimaging, “Omics”, Data Sciences in research centers throughout Europe to develop lithium (7Li) MRI [5-6] to characterize brain lithium distribution and optimize the early prediction of lithium treatment response of newly diagnosed BD patients.
BD patients (37 ± 12 years; 15F/15M) were recruited from across Europe. 3D 7Li MRI were acquired from 3T Siemens or Philips MR scanners equipped with dual-resonance 1H/7Li radiofrequency coils (Rapid Biomedical, Germany) three months after the beginning of their lithium treatment using a bSSFP sequence [6-7] optimized for 7Li signal detection (TE/TR=2.5/5ms, FA 34°, 25mm isotropic resolution). 3D T1-weighted anatomical reference images were obtained using a MPRAGE sequence (1mm isotropic resolution). Comparable data was also acquired at 7T in one centre (not presented here). Our brain [Li] quantification pipeline is schematized in Figure 1. 7Li MRI were interpolated and co-registered to anatomical reference images. To better estimate [Li] in the brain parenchyma and CSF(+eyes) compartments, 7Li MR data were acquired from identical Li reference phantoms (LiCl 2 mmol·L⁻¹ in water/glycerol mixture) for each site and its T1 and T2 relaxation times were estimated. For each center, 7Li signal was calibrated using these identical, batch-produced external references. A partial volume correction (PVC) was applied using the “iterative Yang” method [8] to lessen the substantial signal bleeding from CSF due to the limited spatial resolution of 7Li images. Homogeneous ⁷Li distribution regions were segmented semi-automatically [9] to enable PVC. A correction for the differential T2/T1 weightings was then applied using the bSSFP signal equation [10], the flip angles and reported relaxation times for CSF and brain tissue [6]. 7Li MRI were projected into the MNI-152 space [11-12] to allow region-based analysis. Eight regions of interest (ROIs) were then extracted from the Harvard-Oxford [13-14] and MNI-152 [12] atlases to investigate brain lithium distribution across our cohort. Regional Li concentrations are summarized in Table 1. To account for some of the inter-individual variability (attributable in part to the varying delay between the MRI examination and the last lithium intake), each [Li] map was normalized relative to the average [Li] value in the eyes as a proxy of plasma Li concentration.
Response to lithium was evaluated by a panel of experts applying the Alda scale to clinical ratings acquired prospectively over data 24 months after treatment initiation. This yielded a classification of the BD patients as either good responders (GR, n=11), partial responders (PaR, n=12) or non-responders (NR, n=7). Figure 2 shows the mean brain Li distributions for each “lithium treatment response” group. One can appreciate the apparently higher brain [Li] levels in PaR and NR groups compared to the GR group, in particular for the “relative” Li maps.
Figure 3 presents the “relative” Li concentrations averaged across each ROI for each lithium treatment response groups. An Ordinary Least Squares (OLS) linear regression model was applied to those data with each BD patient’s sex, age and lithium treatment response as regressors. Lithium treatment response was significantly associated with “relative” Li concentrations in most regions (p < 0.05), indicating a consistent influence. In contrast, age and sex showed no significant associations, suggesting the observed effect is independent of demographic factors. Our quantification pipeline incorporated external signal referencing, PVC and differential T2/T1 weighting correction steps to yield coherent [Li] values despite the multicentric nature of our database. Lower apparent brain Li content seems to be predictive of a positive response to lithium treatment. This could be related to differences in Li cellular compartmentation between groups. Indeed, one could expect shorter relaxation times for intracellular or “bound” 7Li. If a larger fraction of 7Li+ were “bound” in GR compared to PaR and NR, it would lead to an underestimation of [Li] levels. Additional 7Li relaxometry or Multiple Quantum Filtering experiments could help ascertain this hypothesis. 7Li MRI remains the only method to investigate brain lithium distribution non-invasively. Additional analysis is ongoing to investigate the relationship between Li brain distribution and clinical, anatomic or metabolic outcomes.
Mariam EL BALQ (Saclay), Gerard HALL, Antoine GRIGIS, Karthik CHARY, Franck MAUCONDUIT, Pete E THELWALL, Aymeric GAUDIN, Marie CHUPIN, Dimitri O PAPADOPOULOS, Letizia SQUARCINA, Thomas SCHULZE, Lars V KESSING, Michael BAUER, Paolo BRAMBILLA, Daniel KEESER, Maj VINBERG, Bruno ETAIN, Philipp RITTER, Edouard DUCHESNAY, Frank BELLIVIER, David A COUSINS, Fawzi BOUMEZBEUR
11:00 - 12:30
#47829 - PG490 Diffusion-weighted MR spectroscopy of the pathological prostate.
PG490 Diffusion-weighted MR spectroscopy of the pathological prostate.
The micro-structure and -environment of tissues can be probed by Diffusion-Weighted MR Spectroscopy (DW-MRS) by simultaneously quantifying apparent diffusion coefficients (ADCs) of metabolites and water [1,2,3]. In a recent study we established DW-MRS of the prostate, providing reference metabolite ADC values for healthy tissue [4]. Prostate pathologies are anticipated to induce microstructural changes detectable by DW-MRS. This work presents the first investigation of metabolic ADCs measured simultaneous with concentration variations across prostate pathologies, including prostate cancer (PCa), prostatitis, and benign prostatic hyperplasia (BPH).
Twenty-five patients with elevated PSA (≥9 ng/mL) underwent a 3T multi-parametric MRI examination. Single-voxel DW-MRS extended the exam by 15 minutes, using a metabolite-cycled STEAM sequence (TE/TM/TR 33/35/2500 ms) and b-values of 124, 776, 1988 s/mm². Voxels targeted low-intensity lesions identified on T2W and ADC images. The voxels were of varying dimensions (3 – 22 cm3) because of differences in lesion size. Clinically suspicious lesions were biopsied under ultrasound or MR guidance, with histopathological analysis. The MRS signals of citrate (Cit), total choline (tCho), spermine (Spe), total creatine (tCr) and water were analyzed. Post-processing involved motion correction, 2D spectral fitting for ADC extraction, and absolute quantification of metabolite concentrations using water signals, with T1/T2 corrections. Quality control involved blinded evaluation by seven spectroscopists.
In healthy controls and a number of patients 3 additional b-values were measured allowing to fit the water signal decay with a slow and fast diffusion component. Among the 25 subjects, 11 were diagnosed with PCa, 7 with prostatitis, and 5 with BPH. Two had both prostatitis and BPH (included in the prostatitis group). Diagnoses were based on biopsy results or radiological reports. Distinct metabolic patterns were observed across pathologies (Fig.1). The ADCs of the luminal space metabolites Cit and Spe increased in pathological prostates (Table 1), and for PCa also that of mI. In contrast the ADC of intracellular metabolite tCho decreased. The tCho concentration increased in pathology (p= 0.03–0.08), but Cit concentration did not change.
We found moderate to strong correlations between the concentrations of Cit, Spe and mI (left column Fig.2A,B,C), There were also strong correlations between their ADCs (right column Fig 2, A,B,C).
In contrast, we found no correlation between the concentrations of tCho and tCr,. However, a strong correlation occured for their ADCs (Fig 2 D).
Contrary to the ADC of luminal metabolites, the ADCs of water tended to decrease in PCa, prostatitis and BPH (Table 1). No correlations were found between the slow or fast ADC components of water and that of the cellular or luminal metabolites. The increased ADCs of luminal metabolites in the pathological prostate indicate environmental changes: e.g.lumen fluid viscosity and/or ion, protein content. The decreased tCho ADC indicate higher cellular density in PCa, prostatitus and BPH (Table 1), corresponding to traditional DWI. Our findings align with glandular component alterations and support tCho as a stronger PCa marker than Cit. The concentration correlations between luminal metabolites are in agreement with the correlation of their levels in expressed prostatic fluid [5] and the correlations between their ADC is anticipated because of their luminal origin. We previously provided evidence that Cit and Spe are associated in prostatic fluid [4,6], which may also contribute to their correlation.
The absence of correlations between the concentrations of tCho and tCr is understandable because of their different biochemical origin, while the correlation of their ADCs most likely is due to a similar cellular environment.
The findings on the ADC of water indicates that it is affected by factors other than that of the metabolites and supports the notion that the slow and fast water ADC components not simply represent cellular and luminal spaces. This study demonstrates that DW-MRS can identify diffusivity properties of metabolites, simultaneous with their tissue concentrations and with the diffusivity of water in the prostate associated with their micro-environment and pathological changes thereof.
Arend HEERSCHAP (Plasmolen, The Netherlands), Angeliki STAMATELATOU, Rudy RIZZO, Kadir SIMSEK, Sjaak VAN ASTEN, Roland KREIS, Tom SCHEENEN
11:00 - 12:30
#46635 - PG491 Dynamic of total Glutamate and Glutamine in response to a short heat pain stimulus.
PG491 Dynamic of total Glutamate and Glutamine in response to a short heat pain stimulus.
Proton magnetic resonance spectroscopy (MRS) allows noninvasive study of metabolism in vivo. Spectra are acquired usually for at least 2-3 minutes to obtain a sufficient signal. Results acquired from these studies are interpreted from metabolic and neuromodulation points. But if one uses short (a few seconds) stimuli metabolic changes should not be presented. In this case, the reason for an increase in neurotransmitter levels may be the transition of neurotransmitters from a vesicle into free space [1, 2]. It was shown that both major neurotransmitter levels increase in a short period after start of presentation of visual stimulation (1-3s) with consequtive decrease to initial values. Mechanism of neurotransmitter vesicular cycling should be same across different brain regions, therefore the aim of this work is to study the kinetics of excitatory neurotransmitter levels after a short-term heat pain stimulus.
All fMRI images and MR spectra were acquired on a Philips Achieva dStream 3 T MR scanner. Twenty-six healthy subjects took part in the study. Before the study, the subjects reported that they had no major medical, neurological, or chronic pain disorders, and did not take painkillers. The subjects were familiarized with all procedures that were applied to them during the study, as well as the MRI protocol. They signed a written informed consent. Thermal stimulation was performed using a MEDOC TSA-2 with a TSA 16x16 mm thermopad, which was attached to the subject's right arm close to the elbow. Before the MRI scan, the subject was presented with stimuli of varying temperatures and fixed duration (5s), which he orally had to rate on a scale from 0 to 10. For stimulation during the MRI scan, a temperature was selected that the subject considered painful, but could tolerate. Stimulation was carried out using a heat pain stimulus of selected temperature for three seconds, repeated after a current period (between the stimuli, the subject was presented to a baseline temperature of 35 degrees for 9-11s). Spectra were obtained using the PRESS pulse sequence and was placed inside an insular cortex of the left hemisphere (40 × 15 × 28 mm3, 16.8 ml, TR/TE = 2000/35 ms, NSA = 315). We analyzed the dynamics of metabolite concentrations with a time development of 2 seconds, as well as the BOLD effect, depending on the determination of the width and height of the NAA and Cr resonance lines. After excluding subjects that had bad MRS data (5 subj.) and no activation in the region (5 subj.), analisys of data was conducted on a 16 subjects (26,6±4.5 years, m – 6, f – 10). Average stimulus temperature was 46.9±1.4 degrees Celsius. The average ratio of activation regressors to the constant level over the spectroscopic voxel volume was 0.50±0.16%. Based on the concentration measurement results, a trend towards an increase in Glx is observed at 4s after heat pain explosion (∆Glx = 0.49±0.19, +5±2%; p = 0.02, p adj. = 0.08) comparing with values at 0s. The Glx/Cr value normalized to creatine does not increase statistically significantly (∆Glx/Cr = 0.07±0.03, +3.7±1.6%; p = 0.06, p adj. = 0.08. The widths of the Cr and NAA lines do not change upon activation (minimum p-value non-adj. = 0.15). It is assumed that changes in the levels of neurotransmitters caused by short stimulation probably caused their release from vesicles, and the subsequent decrease is associated with their return to vesicles. Thus, in this study, excitation and inhibition processes in the visual nucleus of the brain were assessed for the first time using a flickering chessboard, by directly measuring the amount of neurotransmitters released from vesicles. The study was carried out with the financial support of the Russian Science Foundation (agreement number 23-13-00011).
Alexey YAKOVLEV (Moscow, Russia), Elena VORONKOVA, Maxim UBLINSKII, Ilya MELNIKOV, Olga BOZHKO, Tolib AKHADOV
11:00 - 12:30
#47360 - PG492 Somatotopic reorganization is induced by targeted proprioceptive training : fMRI surface analysis.
PG492 Somatotopic reorganization is induced by targeted proprioceptive training : fMRI surface analysis.
Motor training, frequently used in medicine or sport, induces functional reorganization in the sensorimotor network. However, the distinct contributions of sensory (proprioception) and motor components in these remodeling processes remain uncertain. This study examined functional remodeling and somatotopic changes in primary sensorimotor areas after targeted proprioceptive training of the leg.
Twenty healthy volunteers underwent daily training for a fortnight, with two MRI sessions conducted before and after the training period. The training involved mechanical vibrations applied to the tendons of knee muscles to activate proprioceptive afferents and to induce illusions of movement in only the non-dominant leg, while the participant remained motionless. During MRI, we mapped somatotopic organization by vibrating six different muscles at the hip, knee, and ankle levels of both the trained and non-trained legs, using an MR-compatible pneumatic vibration device. As the somatotopic representation of the leg spans the interhemispheric and central sulci of the cortex, we used surface-based analysis to enhance the accuracy of cortical localization by reducing signal contamination between the two sides of the same sulcus [1]. Univariate analysis identified brain regions activated during movement illusions and allowed us to quantify overlaps between representations of different leg segments in the somatosensory cortex. We further applied multivariate representational similarity analysis (RSA) to assess changes in the similarity of fMRI response patterns pre- and post-training. Movement illusions evoked by the six vibration conditions elicited activation across a large sensorimotor network [2,3]. Following proprioceptive training, we observed specific somatotopic reorganisation in the contralateral primary sensorimotor cortex: reduced overlap between the knee and ankle representations of the trained leg, and enhanced lateralization of activations. By contrast, the overlap between knee and ankle representations increased in the contralateral primary motor cortex of the untrained leg. These univariate findings were corroborated by RSA, which showed a significant dissimilarity increase between the trained knee and ankle in the contralateral primary sensorimotor cortex after training, while no significant difference was found between the knee and the hip. In only two weeks, proprioceptive training alone was sufficient to refine the somatotopic organization of the sensorimotor cortices, enhancing the distinction between the trained segment and its adjacent segments. This contrasts with active motor training, which typically increases representational overlap. This study underlies the impact of proprioceptive component in brain remodeling supporting the relevance of a pure sensory training for sensorimotor rehabiitation. A clinical extension of this work is currently underway in amputees, aiming to reduce maladaptive brain reorganization and phantom limb pain.
Aurore LAPUYADE-AUFOO (Marseille), Lilia PONSELLE, Raphaëlle SCHLIENGER, Laurent THÉFENNE, Bruno NAZARIAN, Julien SEIN, Jean-Luc ANTON, Anne KAVOUNOUDIAS
11:00 - 12:30
#46937 - PG493 Subcortical Dysregulation in Post-COVID Breathlessness using 7T MRI.
PG493 Subcortical Dysregulation in Post-COVID Breathlessness using 7T MRI.
An estimated 10% to 50% of post-COVID individuals experience breathlessness despite a fraction of severe hospitalised ones showing measurable pulmonary injury. COVID-19 is known to affect the central nervous system (CNS), potentially leading to functional alterations in the processing and perception of respiratory signals.
To investigate these neural changes, we acquired 7 Tesla resting-state functional MRI (rs-fMRI) data from 53 post-COVID patients with varying infection severity and breathlessness levels, despite minimal evidence of pulmonary dysfunction. Using univariate robust regression models followed by FDR correction, we evaluated the predictive value of 153 resting-state functional connectivity (rs-FC) measures across 18 brain regions implicated in the hierarchical inference of respiratory interoception and allostasis. We found that decreased rs-FC between the dorsal periaqueductal gray and posterior insula (dPAG-PoI1), along with increased rs-FC between the basolateral amygdala and dorsal anterior cingulate cortex (BLA-dACC), were significantly associated with higher breathlessness catastrophizing scores. Post-COVID breathlessness was characterised by a broader pattern of increased connectivity within visceromotor structures (beyond BLA-dACC) and decreased connectivity in primary interoceptive regions (beyond dPAG-PoI1), despite not surviving FDR correction.
The contribution of dPAG-PoI1 rs-FC to breathlessness remained significant after adjusting for general anxiety, whereas BLA--dACC did not. A significant interaction effect was identified between ventilator use and dPAG-PoI1 rs-FC, but not with BLA-dACC, such that individuals who had undergone mechanical ventilation exhibited a stronger association between decreased dPAG-PoI1 connectivity and greater breathlessness, whereas this relationship was weaker in non-ventilated individuals. The hypo-connected dPAG-PoI1 rs-FC may represent a pathway of reduced integration of afferent visceral signals within primary interoceptive structures, possibly relating to brainstem damage in severely affected individuals, and that it impairs safety signal learning after recovery.
The hyper-connected BLA-dACC rs-FC may represents an exaggerated efferent anticipatory bias toward adverse respiratory experiences within visceromotor structures, contributing the general anxiety, yet possibly not related to the acute-phase severity. Both pathways may prevent the generative model of gas-exchange in the brain from updating when recovered from the acute-infection. Our findings support a dual-pathway model for post-COVID maladaptive breathlessness: hypoconnectivity within the interoceptive dPAG–PoI1 pathway that prevents safety signal learning and hyperconnectivity within the visceromotor BLA–dACC pathway that prime threat actions in face of normal gas-exchange after recovery.
Lin QIU (Oxford, United Kingdom)
11:00 - 12:30
#47653 - PG494 Acute response of hepatic fat content to consuming fructose or glucose alongside fat in obese and non-obese subjects.
PG494 Acute response of hepatic fat content to consuming fructose or glucose alongside fat in obese and non-obese subjects.
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a growing global health concern characterized by excessive fat accumulation in the liver (hepatic steatosis) in individuals without excessive alcohol consumption. While the exact mechanisms are complex and multifactorial, dietary factors, particularly the consumption of fructose, have been implicated in its development and progression. In previous study[1], we demonstrated that fructose if provided together with high fat load led to accumulation of fat in the liver in non-obese subjects. On the contrary the coadministration of glucose with high fat load did not affect hepatic fat content (HFC). In this study we tested whether such a negative effect of fructose is preserved in obese subjects.
Fourteen non-steatotic, non-obese subjects (HFC < 5.56 %, BMI < 30 kg/m2), and eight obese subjects (BMI ˃ 30 kg/m2) were enrolled in this study. The exclusion criteria were diabetes, excessive alcohol consumption, use of drugs affecting lipid metabolism and triacylglycerols ˃ 4.0 mmol/l, and other serious illnesses.
Each subject underwent two almost identical interventions. In each of these interventions the HFC (HFC-0) was measured by 1H MR spectroscopy (MRS) after overnight fasting. Then, subjects consumed 473 ml of high fat cream (150 g of fat) and fruit tea sweetened with 50 g of glucose. Same sugar was given to them again after 2 and 4 hours. Second MRS for HFC determination was measured six hours after the first dose of sugar (HFC-6). Blood was collected before the first consumption and then at 0.5, 1, 2, 2.5, 3, 4, 4.5, 5, and 6 hours after, and stored at -80 °C for later analysis. In the second intervention, the fructose was consumed instead of glucose. The order of interventions was randomized.
The MRS examination was performed in the supine position during held exhalation at 3T MR system (Siemens, Germany) equipped with 8- or 30-channel surface coil and 32-channel spine coil, with use of single voxel spectroscopy sequence (TR = 4500 ms, TEs = 20-33-50-68-80-100-135-150-180-270 ms, 2 acquisitions, water suppression). The position of VOI (40×30×25 mm) was placed in the liver segment V/VIII in the area without visible big vessels. Automatic and manual shimming were combined to reach a linewidth <50 Hz. MR spectra were evaluated by LCModel[2] and the concentrations were corrected for T2 relaxation times in each subject using MATLAB software. HFC was calculated from fat fraction using Longo correction[3]. Relative content of saturated fat fraction in the VOI was estimated from fractions of hydrogen atoms of functional groups[4]. Basic characteristic of both groups is stated in Table 1. Compared to the non-obese group, obese subjects showed increased HFC and higher insulin resistance evaluated as HOMA-IR, and decreased relative lipid saturation.
The relative HFC change (HFC-6/HFC-0 (%)) after fat with fructose was higher in non-obese than in obese subjects. Moreover, in non-obese group, the relative HFC was higher after fat administration with fructose than after fat with glucose (Table 2) contrary to obese subjects, where no statistically significant difference between the glucose or fructose experiments was detected. Insulin level was increased after high fat load with glucose than with fructose (Figure 1) in both groups. No significant changes were found in FFA nor TG in either group or between the two interventions. In this study we observed that HFC in non-obese subjects increased after high fat load with fructose, but not after high fat with glucose. On the contrary, we were unable to detect any change in HFC in the same experiments in obese individuals. The negative impact of fructose coadministration in non-obese subjects can be explained by differences in metabolism of both sugars. Fructose is completely metabolized in the liver and there is no feedback inhibition of fructolysis leading to overproduction of substrate for de novo lipogenesis (DNL). On the other hand, only 20% of glucose is metabolized in the liver and metabolism of glucose is under strict control. The lack of significant changes in circulating FFA and TG between interventions in both groups suggests that the observed HFC changes might be driven by rapid intrahepatic DNL rather than increased peripheral lipid delivery. No observed changes in HFC after fructose consumption in obese subjects might be explained by already increased DNL. It remains to be determined whether insulin resistance typical for obesity plays any role in observed effects. The role of fructose in MASLD pathogenesis is currently under comprehensive discussion and our data may contribute to understanding its role in accumulation of liver fat. Contrary to non-obese subjects, no differences could be detected in the response of HFC to glucose and fructose coadministration with high fat load in obese individuals.
Petr KORDAC (Prague, Czech Republic), Dita PAJUELO, Milan HAJEK, Petr SEDIVY, Monika DEZORTOVA, Petra STASTNA, Jan KOVAR
11:00 - 12:30
#47734 - PG495 COVID-19: Investigation of BC007 for the treatment of patients with Post-COVID syndrome (PCS) using comprehensive CEST imaging.
PG495 COVID-19: Investigation of BC007 for the treatment of patients with Post-COVID syndrome (PCS) using comprehensive CEST imaging.
Patients suffering from Post-COVID Syndrome (PCS), present a wide range of symptoms that are not entirely specific to PCS. To the state of this abstract, no treatment has emerged beyond the status of a clinical study. Initial findings from an internal study (discover 1.0) [1] indicate the need of further subclassification of PCS patients into (i) virus-induced autoimmune reactions, (ii) prolonged recovery due to organ damage with functional impairment, and (iii) sustained immune activation caused by persistent viral components [2]. However, these categories may not fully capture the complex heterogeneity of PCS, highlighting the urgent need for deeper phenotyping and mechanistic insights, especially in the diagnostics
Recent data [3] suggest that a subgroup of patients with PCS has functional autoantibodies directed against a G-protein-coupled receptor (GPCR-fAAb). The neutralization of fAAb by a new drug called Rovunaptabin (BC007) has been demonstrated [4] to ameliorate the symptoms of PCS in a clinical trial. In this randomized controlled study, PCS patients received BC007 and placebo in a cross over design. We monitored the influence of this drug on the intracellular metabolics using 7T CEST MRI in an unprecedented and unique clinical study setup.
29 patients with PCS were scanned on a MAGNETOM Terra.X 7 Tesla scanner with an 32ch Rx and 8ch Tx head coil. Each patient was scanned three times: Subjects underwent comprehensive Chemical Exchange Saturation Transfer (cCEST) imaging [5] with 3D snapshot readout [6] at B1=1, 2 and 4µT to detect slow, intermediate and fast exchanging protons groups from -NH, -NH2 and -OH compounds. PCS patients received up to three measurements with a 2-week interval. While the first measurement can be interpreted as metabolic reference scan before treatment, the patients received in the subsequent scans either a drug treatment with BC007 or a placebo.
For post-processing, cCEST data were B0 and B1. Lorentz amplitude maps from data acquired with B1= 1µT were predicted by a deepCEST network [7] to generate aliphatic NOE, semi-solid MT, amide and guanidine maps. MTRasym maps from the amine and hydroxyl groups were calculated separately (Fig.1). In a first experiment, white matter (WM), grey matter (GM) and cerebrospinal fluid (CSF) were segmented for each patient and the timepoint of the first measurement was compared with age-matched healthy control datasets from our cCEST database. The data were unblinded and analyzed longitudinally in order to investigate if molecular changes appear in cCEST imaging through drug treatment. The segmented white and gray matter revealed a significantly increased amine and hydroxyl signal in the PCS cohort. In contrast, the other CEST markers, as well as T1, did not show significant differences compared to the healthy control cohort (Fig. 2). When observing PCS patients after the intake of placebo or BC007 (Verum), no significant reductions were observed within the WM and GM for the NH2 and OH groups (Fig.3) These data show clearly increased CEST signals of the NH2 and OH groups of patients with PCS compared to age-matched healthy subjects. The intake of BC007 seems to reduce these CEST signals, but cannot be significantly confirmed. This may be related to the small number of participants, as only 19 of the initial 29 PCS patients participated in all 3 measurements. cCEST reveals metabolic changes of PCS patients compared to age-matched healthy controls.While a global evaluation shows differences, especially in amines, more detailed analyses of the WM/GM are necessary in future.
Jan SCHÜRE (Nürnberg, Germany), Moritz FABIAN, Bettina HOHBERGER, Anja-Maria LADEK, Marion GANSLMAYER, Thomas HARRER, Friedrich KRUSE, Stefanie MAAS, Tobias BORST, Ralph HEIMKE-BRINCK, Andreas STOG, Thomas KNAUER, Eva RÜHL, Victoria ZEISBERG, Adam SKORNIA, Armin STRÖBEL, Monika WYTOPIL, Caroline MERKE, Sophia Hofmann SOPHIA HOFMANN, Katja G. SCHMIDT, Petra LAKATOS, Julia SCHOTTENHAMM, Martin HERRMANN, Christian MARDIN, Andy HESS, Jürgen RECH, Armin NAGEL, Arnd DÖRFLER, Moritz ZAISS
11:00 - 12:30
#47762 - PG496 ¹H-MRS Study of Glutamate–Glutamine Dynamics in the Insular Cortex Following Short-Term Heat Pain.
PG496 ¹H-MRS Study of Glutamate–Glutamine Dynamics in the Insular Cortex Following Short-Term Heat Pain.
Functional magnetic resonance spectroscopy (fMRS) is used to investigate task-related changes in brain metabolism and neurotransmitter activity in vivo. Typically, fMRS requires at least 30 seconds of spectral acquisition to achieve sufficient signal-to-noise ratio. Changes observed over these time frames are generally interpreted from metabolic and neurochemical perspectives. However, at shorter time scales, metabolic processes are unlikely to produce MRS-detectable changes[1, 2]. In such cases, transient increases in neurotransmitter levels may instead reflect the rapid release of neurotransmitters from synaptic vesicles into the extracellular space[3]. Previous studies have shown that levels of major neurotransmitters can rise within 1–3 seconds of visual stimulation onset, followed by a return to baseline[4, 5]. Given that the mechanism of vesicular cycling is conserved across brain regions, the aim of this study was to investigate the kinetics of excitatory neurotransmitter levels in the insular cortex following a brief heat pain stimulus.
All fMRI and MRS data were acquired using a Philips Achieva dStream 3T MR scanner. Twenty-six healthy volunteers participated in the study. Prior to participation, all subjects confirmed they had no major medical, neurological, or chronic pain conditions and were not taking pain medications. Participants were fully briefed on all procedures and the MRI protocol and provided written informed consent. Thermal stimulation was presented using a MEDOC TSA-2 system with a 16×16 mm thermopad attached to the right forearm near the elbow. Before scanning, heat stimuli of varying temperatures (5-second duration) was presented to each subject. They rated the pain intensity orally on a scale from 0 to 10. For in-scanner stimulation, a temperature that was rated as painful but tolerable was selected individually for each subject. During the scan, thermal stimulation consisted of a 3-second heat pulse followed by a rest period with baseline temperature (35°C) lasting 7–9 seconds. Stimulus was presented 60 times during MRS and 30 times during fMRI. Spectra were acquired using a PRESS pulse sequence (TR/TE = 2000/35 ms, NSA = 315) with the voxel placed in the left insular cortex (40 × 15 × 28 mm³; volume: 16.8 mL). Spectra were acquired twice: without stimulation (‘sham’ condition) and with (‘act’ condition). In addition, we assessed potential BOLD-related effects by monitoring changes in the width and amplitude of the N-acetylaspartate (NAA) and creatine (Cr) resonance lines. Spectra were preprocessed using FID-A, including phase and frequency alignment, signal averaging, and water suppression. Quantification of metabolite concentrations was performed using LCModel. Metabolite dynamics were analyzed across five time points with a temporal resolution of 2 seconds; the sixth time point was excluded due to a low number of signal averages (NSA). After excluding subjects with poor-quality MRS data (n = 5) and those without significant fMRI-measured activation in the target region (n = 5), data analysis was conducted on 16 participants (age: 26.6 ± 4.5 years; 6 males, 10 females). The average temperature of the pain stimulus was 46.9 ± 1.4 °C. The mean activation regressor (according to fMRI) ratio relative to the constant level within the spectroscopic voxel was 0.50 ± 0.16%. A trend toward increased Glx concentration was observed at 4 seconds after the onset of the heat pain stimulus compared to baseline (∆Glx = 0.49 ± 0.19, +5 ± 2%; p = 0.02, p_adj = 0.08) and the Glx/Cr ratio showed a nonsignificant increase (∆Glx/Cr = 0.07 ± 0.03, +3.7 ± 1.6%; p = 0.06, p_adj = 0.08) in act condition. No changes Glx and Glx/Cr observed during sham condition. No significant changes were observed in the linewidths of the Cr and NAA resonance peaks (minimum uncorrected p = 0.15). fMRI results confirmed activation in the insular cortex, where Glx dynamics were measured. However, compared to visual stimulation paradigms, the average BOLD signal change following the heat pain stimulus was smaller. This reduced BOLD response may explain the more modest increase in Glx levels observed in this study. A lower concentration of Glu release from vesicles likely reflects weaker neuronal activation, which corresponds to the smaller BOLD signal in the region. Additionally, a lower BOLD response may limit the detectability of MRS-BOLD–related spectral changes. The mechanism of vesicular neurotransmitter release may account for a transient increase in Glx following a brief stimulus. However, reliable detection of such changes using fMRS appears to require sufficiently strong neuronal activation.
Alexey YAKOVLEV (Moscow, Russia), Elena VORONKOVA, Maxim UBLINSKIY, Olga BOZHKO, Tolibjon AKHADOV
11:00 - 12:30
#47776 - PG497 Measuring Renal Reabsorption Dynamics with Dynamic Glucose Enhanced (DGE) MRI.
PG497 Measuring Renal Reabsorption Dynamics with Dynamic Glucose Enhanced (DGE) MRI.
Dynamic glucose-enhanced (DGE) MRI utilizes chemical exchange saturation transfer (CEST) [1] or direct water saturation [2] to measure glucose uptake in vivo. In kidneys, this potentially allows for observing glucose reabsorption dynamics, unlike DCE MRI, which can only measure the filtration of the respective contrast agent.
DGE MRI protocols usually require high temporal resolution. To achieve this, often no full Z-spectrum is acquired. Additionally, traditional CEST quantification methods are confounded by magnetization transfer (MT), relaxation effects, and field inhomogeneities.
To overcome these limitations, we aim to utilize model-based analysis techniques to achieve sufficient temporal resolution, while correcting for confounding factors. Model-based approaches have demonstrated to be able to quantify MT and CEST effects in preclinical research [3,4]. We adapted these approaches and developed them further to reliably quantify glucose under dynamic conditions.
All measurements were performed with a 35 mm resonator on a 9.4 T small animal Bruker Biospec MR system. A glucose bolus of 6 μmol/g body weight was injected intravenously into one representative wild-type mouse after 0, 30, and 60 minutes. During the infusion protocol, 20 consecutive CEST measurements were conducted: one axial slice of the left kidney, RARE sequence, 32x32 matrix, 0.2x0.2 mm^2 resolution, 27 offsets, B_1=1.6 μT, 2s saturation, at least 2s recovery, with respiration trigger applied. The total acquisition time per spectrum was approximately 4 min. All data were denoised with a total-variation denoising algorithm [5].
For the model-based analysis of CEST MRI data, we simplified the steady-state solution for Z-spectra by Zaiss et al. [3] assuming that glucose exchange rates are much larger than the nutation frequency ω_1 and that for Δω≲ω_1, the MT relaxation effects are dominated by the direct water saturation:
Z_ss (Δω)=(R_(1,obs)⋅Δω^2)/((R_(1,obs)+f_MT/(1+f_MT )⋅R_rf^MT (Δω))⋅Δω^2+(A⋅Δω+B)⋅ω_1^2 ),
where R_rf^MT (Δω) is the MT absorption line-shape function. Here, we assume a Lorentzian line-shape:
R_rf^MT (Δω)=(ω_1^2⋅R_(2,MT))/(R_(2,MT)^2+(Δω-δω_MT )^2).
R_(1,obs) was provided from an additionally measured T_1-map. B_0- and B_1-maps were obtained using WASABI [6] and used for explicit correction of Δω and ω_1. Fitting was performed in two steps: all parameters were fitted to the mean spectrum of one kidney compartment (pelvis, medulla, cortex). The obtained δω_MT was then fixed and A,B,f_MT,R_(2,MT) were fitted to the pixelwise spectra with initial values set to results of the first fitting step. Figure 1 shows mean Z-spectra, MTRasym and AREX curves for different kidney compartments, as well as fit results for each region before the first glucose infusion (baseline). Residuals of the fits oscillate closely around zero, indicating most of the spectral asymmetry was captured by the model.
Baseline maps of MTRasym, AREX, and asymmetry parameter A (Figure 2) revealed substantial differences in the calculated CEST contrasts. For MTRasym the values in the pelvis were relatively larger compared to other compartments, while AREX showed a mostly homogeneous distribution across the whole kidney and model-based analysis showed higher asymmetry in the cortex.
Figure 3 depicts snapshots of the difference between MTRasym, AREX, A and their corresponding baseline values throughout DGE MRI. Compared to the two metrics, the model parameter map shows a much smoother, less fractured pattern and clear glucose accumulation in the cortex.
Mean time courses of ΔMTRasym, ΔAREX, and ΔA for different kidney compartments revealed glucose signal peaks following each infusion time point (Figure 4). AREX and A showed higher signal difference in the cortex compared to medulla and pelvis. However, this effect was much more pronounced for the model-derived contrast. The calculated DGE contrast strongly varied with the chosen analysis method. MTRasym does not account for T1- or T2-relaxation effects, which lead to an over-estimation of the asymmetry in the pelvis. AREX considers T1 effects but offers no explicit B_0- and B_1-correction and does not account for T2 related water saturation broadening and asymmetric MT effects.
Only the model-based approach considers all these effects, which lead to much high glucose signal observed in the cortex, where the reabsorbed glucose should remain. This indicates that model-based analysis offers better access to the true underlying glucose dynamics in healthy kidneys. The presented simplified model for steady-state Z-spectra remained stable to fit even at pixelwise noise levels.
Following up on this research, the temporal resolution might be improved using shorter saturation (Spin-Lock) and Gradient-Echo snapshot acquisition. In this study, DGE MRI in combination with model-based analysis was demonstrated to detect glucose reabsorption dynamics in the kidney. In future studies, this might offer novel insights into renal pathologies.
Chris LIPPE (Münster, Germany), Verena HOERR
11:00 - 12:30
#47783 - PG498 Resting-state fMRI signals associated with EEG-detected eye blinks during multimodal acquisitions.
PG498 Resting-state fMRI signals associated with EEG-detected eye blinks during multimodal acquisitions.
There is a growing interest in spontaneous fluctuations of functional brain networks during rest measured using fMRI, EEG, or their combination [1]. Eye blinks, linked to vigilance and arousal processes that contribute to these fluctuations [2], have been associated with activity surges within the ascending arousal network [3], indicating their potential as markers of arousal events. Such events have been further linked to large-scale cortical BOLD signal fluctuations and pulsatile cerebrospinal fluid (CSF) flow [4,5]. Importantly, arousal events can vary in intensity and duration, with higher-intensity arousals often eliciting stronger physiological responses [6]. Eye blinks during rest also vary in frequency, resulting in differing pre-blink intervals that may reflect distinct types of arousal events. They are typically detected using eye tracking; however, in the absence of such recordings, they can also be extracted from the EEG signals. In this study, for the first time, we detect eye blinks based on the EEG signals recorded simultaneously with fMRI, and use the EEG-detected blinks as arousal event markers, investigating their fMRI correlates, including cortical and subcortical brain regions as well as global and CSF signals.
Simultaneous EEG-fMRI was acquired from 45 participants (9 M, 36 F; age 29.8 ± 7.6 years) during 7min (n=29) or 10min (n=16) eyes-open resting state sessions. EEG data were recorded with a 32-channel MR-compatible system (Brain Products) at a 5 kHz sampling rate. EEG signals were corrected for gradient and pulse artefacts and bandpass filtered. Eye blinks were detected using the BLINKER pipeline applied to frontal EEG channels [7], with those occurring within one 1 TR (1.26 s) merged. Blinks were then categorised into two groups based on the pre-blink interval: (i) clustered blinks with at least one prior blink within the preceding 5 seconds and (ii) isolated blinks with no prior blink activity for at least 10 seconds. The EEG data were then further preprocessed and used to extract a scan-level vigilance index given by the ratio of the averages of the alpha to delta and theta powers. Subjects were divided into low- and high-vigilance groups based on the 40% and 60% percentiles of the vigilance index distribution. fMRI data were collected on a 3T MRI system (Siemens) using a 64-channel head coil. T2*-weighted multi-slice GRE-EPI sequences were employed (TR/TE = 1260/30 ms, GRAPPA = 2, SMS = 3, 2.2mm isotropic resolution). The data were analysed using FSL tools, and pre-processing included motion and distortion correction, high-pass temporal filtering and spatial smoothing. Voxel-wise general linear model analyses were performed using two blink time series, corresponding to the clustered and isolated blink groups, as regressors of interest. Analyses were conducted across a range of time lags (-10 to +10 TR, with a 1 TR step) to capture temporal dynamics. Motion parameters and outliers were included as nuisance regressors. A group-level mixed-effects voxelwise analysis determined the main effects of blinks on the fMRI signal across the brain in each group using cluster statistical inference. A region-of-interest (ROI) analysis was performed by averaging the resulting voxelwise Z-scores, including whole brain and CSF (lateral and 4th ventricles) as well as cortical and subcortical regions. Fig. 1 shows the distribution of the normalised blink counts for clustered and isolated blinks for low and high vigilance subjects. Voxel-wise fMRI analyses (Fig. 2–3) revealed pre-blink activations in subcortical and visual cortical regions, more pronounced during low vigilance scans, followed by widespread cortical deactivation, particularly evident in high vigilance. These patterns are consistent with previous findings based on eye tracking. Isolated blinks were associated with a delayed reduction in cortical BOLD signal when compared to clustered blinks. ROI-based analyses (Fig. 4) corroborated these results. Additionally, blink-related signal modulations were observed in the fourth ventricle, more prominent following isolated blinks, suggesting a potential link to arousal-related CSF dynamics. These findings support the use of EEG-detected eye blinks as markers of arousal events during resting state fMRI, in line with prior eye tracking research [3]. Crucially, the distinction between clustered and isolated blinks leads to differences in both the timing and amplitude of associated BOLD signal changes, indicating heterogeneity among arousal-related events. The observed modulation in the fourth ventricle signal adds to the emerging body of evidence linking neural activity to CSF flow dynamics in awake humans [4,5]. The study shows that EEG-detected blinks may be effective arousal markers in resting-state fMRI. Differences between clustered and isolated blinks reflect distinct BOLD responses, highlighting arousal-related variability and linking blink timing to neural and CSF dynamics.
Frederico SANTIAGO (Lisbon, Portugal), Inês ESTEVES, Ana FOUTO, Amparo RUIZ-TAGLE, Gina CAETANO, Patrícia FIGUEIREDO
11:00 - 12:30
#47843 - PG499 Olfactory functional MRI: Application to evaluate brain activation patterns in women with sexual interest-arousal disorders.
PG499 Olfactory functional MRI: Application to evaluate brain activation patterns in women with sexual interest-arousal disorders.
Olfactory functions such as odor detection or response to olfactory stimuli can be assessed by functional MRI and contribute to the understanding of brain and behavior under different conditions. However, in comparison to other sensory domains, fMRI under olfactory stimulation has been less explored, in part due to the need for specific olfactory delivery systems. We performed olfactory-based fMRI using a custom-built system and applied it to evaluate the response to pheromone in female sexual dysfunction (FSD) subjects before and after treatment. FSD significantly impacts quality of life, with the most common subtype being hypoactive sexual desire disorder (FSIAD), characterized by a persistent lack of sexual desire or fantasies. Its causes are not fully understood but likely involve neuroendocrine, psychiatric, and behavioral factors (1,2). Only two studies have directly compared brain activity in women with and without FSIAD (3, 4). This study hypothesizes that FSD and the improvement in sexual dysfunction observed with ospemifene treatment is associated to changes in the response to pheromone, that can be measurable through olfactory stimulus-based fMRI (5).
We conducted fMRI analyses to study brain activation patterns before and after ospemifene treatment, in a cohort of 15 women with FSD (9 with ospemifene treatment, 6 with placebo). Participants were exposed to alternating scents—pheromones, clean air, and Phenyl Ethyl Alcohol (PEA) —using an open-source, low-cost, custom-built olfactory delivery system compatible with the fMRI setting. The fMRI protocol followed a standard block design with 12 one-minute alternating scent exposures. Head movement was minimized using individually molded foam supports. Prior to functional imaging, a high-resolution T1-weighted structural scan was acquired to exclude anatomical abnormalities and enable accurate localization of brain activity. Functional images were preprocessed with motion correction, EPI correction with T1 and a registration to the MNI152 atlas (6). First level analysis was performed afterwards, to obtain each subject activation in response to each odor in comparison with clean air, followed by a dual regression and randomization. The activation before and after treatment in each treatment group (ospemifene and placebo) was compared using voxel-wise randomize with TFCE correction. Significance was defined as p<0.005. Fig 1.A shows the mean activation map in response to pheromone and PEA, showing a similar pattern of activation in areas related to olfaction, but with some differences in more occipital regions. Regarding the treatment effect, in women who received ospemifene, decreased brain activation was observed after treatment in response to both pheromone (fig.1B) and PEA odors (Fig 1C). Specifically, changes were noted in the frontal pole and medial frontal cortex for the pheromone, and in the insula, parietal operculum, and planum temporal for PEA. In the placebo group, changes between pre- and post-treatment were smaller. Reduced activation to pheromone was seen in the frontal pole (compared to no stimulus) and in both the frontal pole and cingulate gyrus (compared to PEA). This study demonstrates that olfactory-based fMRI can detect changes in brain activation associated with sexual dysfunction and its treatment. Specifically, we observed a significant reduction in neural response to both pheromone and PEA stimuli following ospemifene treatment, particularly in brain areas involved in emotional processing, attention, and sensory integration, such as the frontal pole, medial frontal cortex, and insula. These changes were not as prominent in the placebo group, suggesting a potential effect of ospemifene on olfactory-related brain processing. Although previous studies on FSIAD have been limited, our findings support the hypothesis that olfactory responses, and possibly underlying limbic and cognitive pathways, are modulated by pharmacological intervention in women with FSD. This supports a broader neurobiological involvement in sexual desire disorders, beyond hormonal or psychological explanations. In conclusion, our results suggest that fMRI combined with olfactory stimulation is a promising tool to investigate the neural mechanisms underlying female sexual dysfunction and its treatment. The use of a low-cost, open-source olfactory delivery system demonstrates the feasibility of incorporating such approaches in clinical research. Ospemifene treatment was associated with measurable changes in brain activation, supporting its potential to modulate neural circuits involved in sexual function. Further studies with larger cohorts and longitudinal designs are warranted to validate these findings and explore their clinical significance
Iñigo HERRERO VIDAURRE (Barcelona, Spain), Ribera-Torres LAURA, Jorge OTERO, Ramon FARRÉ, Camil CASTELO-BRANCO, Emma MUÑOZ-MORENO
11:00 - 12:30
#47839 - PG500 Optimization of glutamate quantification using CEST-MRI for neuronal compartment imaging.
PG500 Optimization of glutamate quantification using CEST-MRI for neuronal compartment imaging.
GluCEST imaging has been proposed to image brain glutamate distribution with a better resolution than spectroscopic methods and has many potential applications for the study of neurodegenerative diseases [1]. In this study, we pushed further the limits of gluCEST imaging by combining high magnetic field and high performance cryoprobe to acquire gluCEST images with the best resolution so far.
To demonstrate the potential of quantitative gluCEST mapping, we focused on mouse hippocampus, a highly organized structure composed by several neuron-rich layers with high concentrations of glutamatergic synapses. Whereas gluCEST enables in vivo mapping of glutamate distribution, quantification remains challenging [2,3]. To overcome this obstacle, we developed a signal-optimization pipeline to improve the quantification of glutamate in vivo at 11.7T based on our 6-pools model [4].
CEST images were acquired in 2 anesthetized mice at 11.7T (Bruker) using a cryoprobe and a RARE sequence (B1 = 5 μT, Tsat = 1 s, saturation offsets = [-5:0.2:5] ppm, 0.15 x 0.15 x 0.3 mm3 resolution). A WASSR map was acquired for B0 correction. A B1 map was acquired using the double-angle method to correct for field inhomogeneities. Raw data were denoised using multilinear singular value decomposition (MLSVD) [5] and then fitted using our 6-pools model [4] (creatine (Cr), glutamate (Glu), amide (APT), nuclear overhauser effect (NOE), magnetization transfer (MT) and myo-inositol (MI)). Hippocampal correlation maps, mutual-information maps, and homogeneous-parameter maps were generated to identify the most interdependent parameter pairs to be fixed. Fit quality was assessed by RMSE, the coefficient of determination (R2), the Akaike Information Criterion (AIC), its corrected analogue for small sample sizes (cAIC), and the Bayesian Information Criterion (BIC) calculated as follows:
BIC=nlog(σ^2 )+klog(n),
AIC= nlog(σ^2 )+2k,
cAIC=AIC+ (2k(k+1))/(n-k-1),
Where n is the number of offsets and k the number of degrees of freedom. MLSVD preprocessing increased AIC, BIC and cAIC of the hippocampal Z-spectra by 10.8 %, 7.6 % and 11.1 % respectively (Fig.1). Strong interdependence (r>0.4) among [Cr], [APT], and [MT] was revealed by the correlation maps (Fig.2a) and mutual-information maps (Fig.2b), and confirmed on homogeneous-parameter histograms (Fig.2c), including water-shift: voxel histograms displayed Gaussian distributions. [MT], [APT], [Cr] and water-shift were fixed at 8633 mM, 1998 mM, 86 mM and 0, respectively. At the same selected hippocampal voxel as Fig.1, statistical criteria were estimated to compare fitting performance based-on on raw data (Fig.3c), MLSVD-denoised data (Fig.3d), and MLSVD-denoised data and B1 correction (Fig.3f). MLSVD denoised improved AIC, BIC and cAIC by 12 %, 10 % and 12.1 %, respectively; with both MLSVD and B1 correction, fit criteria were improved by 12.9 %, 10 % and 12.9 %, respectively. In both cases, RMSE decreased from 0.008 to 0.005 and R2 increased from 0.979 to 0.992. The quality of quantitative gluCEST map was significantly improved compared to standard MTRasym map (Fig.4). The solid line (gluCEST, σ = 0.08) is markedly narrower than the dashed line (MTRasym, σ = 0.17), quantitatively confirming its reduced relative dispersion. Enhanced fit quality was observed in the hippocampus, with all statistical criteria showing improvement (Fig.4). The selection of fixed parameters guided by correlation and mutual-information maps ensured the coherence of homogeneous-parameter maps, as reflected by their gaussian voxel-value distributions. Furthermore, signal quality in ventral regions was significantly improved in spite of high B1-heterogeneity inherent to the transmission-reception cryoprobe (Fig.3c). The signal-optimization pipeline associated Z-spectrum fitting based on our 6-pools model significantly enhanced the quality of in vivo gluCEST parametric maps in the mouse brain at 11.7T, and particularly in the hippocampus. We observed higher glutamate concentrations in the CA1 and dentate gyrus regions of the hippocampus which is consistent with higher concentration of glutamatergic neurons in these particular substructures [6]. This quantification tool, when integrated with ultra-high-resolution imaging, holds the potential to enable highly specific visualization of the neuronal compartment. Such an approach could significantly enhance our understanding of the pathological mechanisms underlying neurodegenerative diseases, particularly Alzheimer’s disease, which is known to selectively affect neuronal structures.
Pierre LEMOIS (Paris), Julien FLAMENT
11:00 - 12:30
#46460 - PG501 Role of intramuscular inorganic phosphate in liver transplant candidates.
PG501 Role of intramuscular inorganic phosphate in liver transplant candidates.
Sarcopenia is well recognized in elderly individuals but is also highly prevalent among patients with chronic diseases, including end-stage liver disease, where the prevalence is approximately 40%. It negatively affects patient status before transplantation and significantly influences post-transplant recovery and survival.
This study aims to describe the physical condition and metabolic status of calf muscles of patients before liver transplantation and wants to check these hypotheses:
1) there exists a significant difference in 31P MRS profiles of patients and controls;
2) MRS parameters correlate with simple clinical and kinesiological parameters as are e.g. liver frailty index (LFI) [1], 6-minute walk test (6MWT), etc.
In total, 51 patients (f/m=16/35, mean age 57.4±8.8 y/o, mean MELD score = 18.04) with liver cirrhosis of alcohol etiology were examined at the time of their inclusion on the liver transplant waiting list. The results were compared to those of 22 healthy volunteers (f/m=10/12, mean age 56.3±8.7 y/o). Patients, as well as volunteers, underwent kinesiological tests used for LFI calculation, 6MWT, and MRI and 31P MRS examination of the calf muscles. All subjects gave their written informed consent before participating in the study.
31P MR spectroscopy was performed at 3T MR system VIDA (Siemens Healthineers, Germany) using flexi 31P/1H surface coil (Rapid Biomedical, Germany) underneath calf muscle. Two FID sequences were applied (TR=15 s, TE*=0.4 ms, 16 acq at rest; dynamic part: TR=2 s, 1 acq, 420 measurements, 1 minute rest - 4 minutes plantar flexion at 25% maximum voluntary force - 9 minutes recovery). Signal intensities of PCr, Pi, ßATP, PDE and PME arising predominantly from the m. gastrocnemius and soleus were evaluated using the AMARES fitting routine in the jMRUI v5.0 software.
23 parameters from clinical tests, MRIs, and MRS examinations were used to compare both groups. The comparison of control and patient groups was done using t and U tests in PRISM and JMP software. Significant differences were described by False Discovery Rate (FDR, Benjamini-Hochberg), and significant difference between groups was considered for p<0.05. Results of functional and MRS tests are summarized in Table 1 for 21 parameters, and the difference between groups of subjects and patients was found for 8 variables. These results can be summarized as follows:
a) Patients consistently have lower functional measures (e.g., 6MWT, LFI, power output, area of gastrocnemius), confirming reduced strength and mobility in patients.
b) Metabolic and phosphate-related markers (e.g. Pi/sum) show clear differences between groups, confirming altered muscle metabolism. On the contrary, dynamic MRS parameters did not reveal any significant changes.
The most significant changes were visible in kinesiological tests, especially in 6MWT, where patients walked less than half the distance of the controls (see Table 1).
The correlation matrix of above mentioned 21 parameters shows a strong correlation between 6MWT and LFI but a moderate correlation between clinical/kinesiological tests and MRS parameters (see Figure 1). A key consideration in this sarcopenia study is the quality of the MR spectra. While the signal-to-noise ratio (SNR) was sufficient for reliable quantification of PCr, Pi, and ATP, the signal intensities of PME and PDE should be interpreted with caution, as their lower SNR renders them less suitable for detailed analysis.
Expected changes in the decreasing PCr/Pi signal intensity ratio were observed in all patients, confirming their muscle impairment. However, a more detailed analysis revealed that the observed alterations are not primarily due to changes in PCr levels, but rather to an accumulation of Pi. This Pi accumulation supports the hypothesis of calcium phosphate precipitation, as originally proposed by Allen et al. [2]. According to this mechanism, elevated Pi enters the sarcoplasmic reticulum (SR), where it binds to Ca²⁺, forming insoluble calcium phosphate complexes. This leads to a reduction in the free Ca²⁺ concentration within the SR, thereby limiting Ca²⁺ release and contributing to impaired excitation–contraction coupling during muscle fatigue. Recent studies have provided strong experimental and theoretical support for this hypothesis, demonstrating that Pi-induced reductions in SR Ca²⁺ availability are a significant contributor to fatigue-related muscle dysfunction. In conclusion, simple kinesiological tests may be sufficient for characterizing the current physical condition of liver transplant candidates in routine clinical practice. However, MR spectroscopy provides complementary metabolic information about skeletal muscle, with the inorganic phosphate signal emerging as a particularly promising marker for investigating the pathophysiological background of sarcopenia.
Monika DEZORTOVA (Prague, Czech Republic), Petr SEDIVY, Petr KORDAC, Dita PAJUELO, Robert CHARVAT, Pavel TAIMR, Milan HAJEK
11:00 - 12:30
#47664 - PG502 Automated whole liver segmentation to enable efficient hepatic function assessment with constrast-enhanced mri following selective internal radiation therapy for liver cancer patients.
PG502 Automated whole liver segmentation to enable efficient hepatic function assessment with constrast-enhanced mri following selective internal radiation therapy for liver cancer patients.
Hepatic function (HF) evaluation is essential for monitoring chronic liver diseases and liver cancers like hepatocellular carcinoma (HCC) [1]. Clinically, methods such as hepatic scintigraphy, indocyanine green (ICG) clearance test, ALBI grade [2], and MELD score are utilized, though these offer limited insights about anatomy. Hepato-specific MR contrast agents [3,4] provide a less invasive way to visualize liver tissue properties while delivering anatomical information without ionizing radiation. This study introduces an MRI-based approach to assess HF changes in patients with liver cancer undergoing selective internal radiation therapy (SIRT) and compares it to clinically validated methods.
This prospective single-center study was approved by the ethics committee. Sixteen patients with either HCC or liver metastases from other primary cancers treated by SIRT and with an ICG clearance test, dynamic hepatobiliary scintigraphy, lab tests, and a gadoxetic acid–enhanced liver MRI (3T or 1.5T, Siemens or Philips) before and two months after SIRT were included. Detailed information is in Fig. 1.
HF change was quantified by assessing the evolution in healthy liver volume from gadoxetic acid–enhanced MRI, relying on an automatic liver segmentation and a volume of interest (VOI) of size between 0.75–1 cm³ manually placed in the liver parenchyma as illustrated in Fig. 2. Liver segmentations were performed automatically [5] and manually refined by a radiologist. VOI quality was checked via variation coefficient control, by ensuring that <30% of voxels deviated by > one standard deviation from the VOI mean intensity. For each patient, we ensured that the VOI was selected at the same location between the baseline and the post-op scans.
MRI intensity was normalized using the VOI median intensity value for consistency across scans, scanner manufacturer, and timing differences between contrast injection and image acquisition. The VOI also served for the HF evaluation. A thresholding operation was performed to define the healthy liver volume, according to the formula:
S_(liver voxels) ≥ μ_NVOI-n∙std_NVOI (1)
S_(liver voxels) represents normalized signal intensity in liver voxels [6], μ_NVOI and std_NVOI are the median and the standard deviation of the VOI normalized signal intensity, respectively. The tuneable parameter n sets the intensity threshold to distinguish healthy from abnormal tissue (e.g., tumor areas, regions affected by SIRT, cysts). Eventually, the relative change in healthy tissue volume between baseline and the post SIRT MR scans was computed to classify the HF evolution. A Receiver Operating Characteristic (ROC) analysis was used to find the optimal threshold.
ALBI grade was calculated using albumin and bilirubin levels from lab tests [2] and used as ground truth. A threshold for absolute change was set: an ALBI grade increase >0.1 indicates HF decreases. The ICG value obtained at 15 min post injection (ICG15) was used in the analysis. Liver uptake rate obtained by scintigraphy, ICG15 values, and our MRI-based approach were compared to ALBI grade calling upon ROC curves. Since liver uptake rate and ICG15 are already relative values, solely their absolute change was considered to classify HF evolution.
We determined the parameter n based on the largest area under the curve (AUC) shown in Fig. 3(a, b) and selected n = 9. Our MR-based approach showed an improved accuracy compared to scintigraphy in classifying patients with decreased HF from those with an increased or unchanged HF following SIRT (AUC of 0.78 versus 0.6), although still below ICG15 (AUC = 0.82), as shown in Fig. 3(c). The F1 score of our method reached 0.84, close to ICG15's 0.86, and superior to scintigraphy’s 0.63. Accuracy was slightly lower for HCC patients for all the methods as shown in Fig. 4(a). ROC curves could not be computed for metastatic liver disease as a decrease in HF was systematically observed for these patients. However, 80% of patients (4/5) were correctly classified by our method and ICG15, compared to 60% by scintigraphy.
We also evaluated the effect of treatment selectivity on the different methods. As shown in Fig. 4(b, c), we observed an increased accuracy for the ICG15 and MR-based methods while scintigraphy exhibits a decrease in accuracy in terms of AUC when the treatment becomes more selective. ROC curves could not be computed for whole liver treatment as a decrease in HF was systematically observed. Gadoxetic acid–enhanced MRI scans offer a robust mean of assessing HF evolution, surpassing scintigraphy in both F1 score and AUC, and nearing the accuracy of ICG15. These prospectively determined findings highlight the potential of this non-ionizing method to monitor HF changes following SIRT while providing additional relevant anatomic information. Our method also demonstrated its effectiveness across different MRI field strengths. Further larger prospective trials are needed to validate our data.
Mathieu RUCH (Bulle, Switzerland), Chloé AUDIGIER, Flavian TABOTTA, Bénédicte MARÉCHAL, Rafael DURAN
11:00 - 12:30
#46220 - PG503 Detection of Aggressive Mesenchymal Glioblastoma by Mannose-Weighted CEST MRI.
PG503 Detection of Aggressive Mesenchymal Glioblastoma by Mannose-Weighted CEST MRI.
Glioblastoma is one of the most aggressive cancers known to men. Non-invasive assessment of aggressiveness is crucial for treatment planning, but current MRI protocols lack specificity. Amide proton transfer CEST MRI can grade diffuse gliomas, but not GBM aggression levels. GBM invasiveness arises from a shift from a pro-neural to mesenchymal phenotype. Based on a report that mannose-weighted (MANw) CEST MRI can detect unlabeled mesenchymal stem cells (MSCs) overexpressing mannose [1], we investigated if mesenchymal cancer stem cells can be detected “label-free” in a similar fashion.
Mannose expression was assessed using fluorescein-labeled galanthus nivalis lectin (GNL-FITC, specific for mannose) staining, and the mesenchymal cellular phenotype by anti-CD44 immunostaining. A tissue microarray, containing 35 cases of glioblastoma and 5 cases of normal cerebral tissue was obtained from Tissuearray.Com. CD44 expression and mannose levels were calculated by measuring mean fluorescence intensity (MFI) from fluorescence images, with values normalized on a 0–100% scale for comparison across samples. For pre-clinical studies, low aggressive GBM1a and highly aggressive M1123 cells were used throughout. MANw CEST MRI was conducted using a Bruker 11.7T vertical bore spectrometer. For in vivo tumor models, 2E5 M1123 and GBM1a spheres were injected into the striatum of NSG mouse brain. In vivo T2-w and MANw CEST MRI was performed 1, 8 and 16 days after injection. Tumor and brain ROIs were manually drawn based on T2-w images. For M1123 cells, the mannose-binding lectins LMAN1 and LMAN2 were knocked down using liposomal transfection with LMAN 1/2 siRNA, and LMAN1/2 expression was quantified with qRT-PCR. For the tissue microarray, analysis of CD44 expression and mannose levels demonstrated a positive correlation (r=0.65, p=0.0003)(Fig. 2C), indicating a connection between elevated mannose levels and mesenchymal phenotypic transitions in GBM, with the two markers absent in normal brain. Low mannose expression was seen for both 2D cell cultures, but 3D M1123 spheres contained more mannose compared to GBM1a (Fig. 1A). In vitro MANw CEST MRI showed the highest CEST signal for the M1123 3D spheres (Fig. 1B). T2-w MRI showed M1123 cells growing much faster than GBM1a invading across the entire hemisphere on day 16 (Fig. 1C). On day 1, a distinct MANw CEST MRI signal was observed for M1123, but not for GBM1a. Eight and 16-day post-injection follow-up revealed a continuous pronounced MANw CEST signal only for M1123 (Fig. 1D), which correlated with anti-mannose staining (Fig. 1E). The MANw CEST signal of M1123 was significantly higher (>1.8-fold) than GBM1a and host brain for all time points (Fig. 1F). Anti-CD44 immunostaining revealed an abundance of MSCs in M1123, but none in GBM1a. Silencing LMAN1/2 in M1223 cells (Fig. 2A) resulted in a 4-fold reduction of LMAN1/2 and mannose expression (Fig. 2B), which was accompanied by a 10% reduction in MANw CEST MRI signal (Fig. 2C). Changes in glycosylation profiles are an important feature of mesenchymal transitions and offer a potential biomarker for classifying tumor subtypes and monitoring transitions to more aggressive treatment resistant tumors. We showed that mesenchymal GBM cells express higher levels of genes involved in mannose regulation and display significantly higher mannose levels and higher MANw CEST signal than their proneural counterparts. Importantly, these correlations were maintained in patient derived glioma cells and tissues and tumor xenografts. While conventional T2-weighted and Gd-based contrast-enhanced MRI enable visualizing brain tumors, they lack specificity in differentiating tumor subtypes and assessing microenvironmental changes. Integrating MANw CEST MRI with the currently available MRI portfolio, including APTw MRI, could enable a more comprehensive evaluation of GBM. We envision this approach will improve detection of aggressive mesenchymal tumors and their recurrence, refine treatment stratification, enhance the monitoring of therapeutic responses, and aid in optimizing surgical planning. We have developed MANw CEST MRI as a novel method to assess GBM aggressiveness without the need of injecting imaging agents. Once translated, this advancement may decrease the time interval between diagnosis and treatment, increasing patient survival. Since brain tumor patients already undergo routine MRI, our approach can be added to existing MRI protocols without further regulatory approval. Clinical studies are currently in progress.
Behnaz GHAEMI, Hernando LOPEZ-BERTONI, Shreyas KUDDANNAYA, Sophie SALL, John LATERRA, Guanshu LIU, Jeff BULTE (Baltimore, USA)
11:00 - 12:30
#47364 - PG504 Optimal control pulses and MIMOSA for CEST preparation at 7 T.
PG504 Optimal control pulses and MIMOSA for CEST preparation at 7 T.
In UHF systems at 7T, highly inhomogeneous B1 fields pose a major challenge for RF pulses. The MIMOSA approach [2], using alternating circular and elliptical polarization, improves saturation homogeneity for conventional Chemical Exchange Saturation Transfer (CEST) pulses. The newly introduced approach of Optimal Control (OC) pulses for the preparation of CEST effects shows advantages over classical pulse shapes [1,4].
This work investigates whether OC pulses retain their properties and benefit similarly from MIMOSA. This investigation was motivated by the complex superposition of electromagnetic phenomena and the special properties of OC pulses.
In contrast to optimized pulse trains in 1Tx systems, pTx using MIMOSA requires to repeat the same pulse with different modes. Thus, it requires a single efficient OC Pulse, which is B1 independent has been optimized for this purpose. This pulse is then concatenated to a pulse train of arbitrary B1rms, duty cycle (DC) and duration and played out using Pulseq pTx and the hybrid Pulseq-Gradient Echo Sequence [8]. The Pulseq pTx extension [7] now allows us to modulate each pulse individually in different RF modes and thus represent the MIMOSA principle. The resulting OC CEST preparation pulse is shown in Fig 1. The High B1 preparation method of the comprehensive CEST approach [3] was optimized with OC pulses. The B1 maps used for the B1 correction were generated using a DREAM [5] mapping method (Fig 2.). For the B0 correction, a B0 map was created with a WASABI [6] measurement. The evaluation of the CEST maps was carried out with the CEST pipeline of the cCEST [3] approach shortened to the High B1 level, 4µT Hydroxy.
All experiments were performed on a 7T Terra.X VA60 (Siemens Healthineers) scanner and an 8Tx/32Rx head coil (Nova Medical) and under approval of a local ethics committee. The measured subject was a male healthy subject. It can be shown that the use of OC pulses with MIMOSA preparation can improve homogeneity [Fig 3]. The strong signal of the CP mode is only homogeneous in the center [Fig 2]. The areas outside the center have fewer and a less stable signal. The MIMOSA mode, which combines the CP and EP modes, ensures that a homogeneous contrast is achieved across a wider area. OC pulse for CEST preparation provides visible improvements for CEST imaging. This has also already been demonstrated at 7 Tesla [4]. MIMOSA preparation has already been shown to improve B1 inhomogeneities in UHF imaging [2]. The combination of the two technologies improves the homogeneity of the CEST maps . The Pulseq Hybrid approach allows new optimized pulse shapes to be easily tested. This allows OC Pulse to be applied flexibly to different B1rms levels, duty cycles and number of pulses. It is also possible to permanently integrate optimized pulse shapes into CEST sequences. The combination of OC pulses and MIMOSA preparation can visibly improve homogeneity of CEST imaging even further
Acknowledgements:
Thanks to Moritz S. Fabian for providing the cCEST evaluation pipeline.
Funded by IDL@7T, BMBF.
Martin FREUDENSPRUNG (Erlangen, Germany), Clemens STILIANU, Simon WEINMÜLLER, Rudolf STOLLBERGER, Moritz ZAISS
11:00 - 12:30
#46239 - PG505 Deviant loading-induced deformation pattern as a potential marker of discogenic pain: A distinct phenotype of nonspecific low back pain?
PG505 Deviant loading-induced deformation pattern as a potential marker of discogenic pain: A distinct phenotype of nonspecific low back pain?
The diagnosis of nonspecific low back pain (LBP) faces several challenges due to the lack of precise biomarkers [1]. MRI often fails to reveal a clear cause, leading to potential misinterpretation of findings [2].
Increased stress on the intervertebral discs may cause fissures and lead to structural weaknesses of the annulus fibrosus [3]. This weakening may alter the biomechanical properties and introduce micro-instabilities in the motion segment [4]. The deformation of such discs may be elevated and cause pain-signaling from nerve endings in the outer part of the annulus fibrosus.
This study aimed to determine whether disc deformation is linked to annular fissuring and if it has the potential to phenotype/target the pain in patients with nonspecific LBP. The aim was investigated using a novel MRI method to assess disc deformation (Fig 1) [5] with CT and low-pressure discography as references of annular fissuring and pain provocation.
76 intervertebral discs in 28 LBP patients [45±9 years, 16 women] were examined with MRI in supine position with and without spinal loading, followed by low-pressure discography and CT during the same day.
To determine the disc deformation, MR images with and without loading were registered to a common spatial volume using the Elastix software. Disc compression or expansion was then calculated as the Jacobian determinant of the registration deformation field in five midsagittal slices (Fig 1). A senior radiologist classified the discs on post-CT-discograms into no fissures, posterior fissures, anterior and posterior fissures, and severe fissuring (<50% continuously intact outer third annulus fibrosus). The operational criteria for pain provocation during discography [6] were applied to classify discs into discogenic pain/no pain.
Wilcoxon rank-sum and Levene’s test were used with p<0.05 to determine group differences and equality of variances. A binary logistic regression model was used to explore possible associations between deformation and fissure extent or pain, where the strength of the model was displayed using receiver operating characteristics (ROC). In general, the intravertebral discs displayed compression of the annulus fibrosus posteriorly and expansion anteriorly (Fig 2) and only small differences within groups and between groups, were seen. However, discs with fissures both anteriorly and posteriorly and those with severe fissuring displayed larger deformation and significantly larger within-group variance (p=0.001-0.04).
Discs with high anterior and low posterior deformation were more likely pain-signaling, whereas those with high posterior and low anterior deformation were more likely non-pain-signaling (Fig 3). These specific deformation patterns could predict pain and no pain in the discs with high certainty (Fig 4). Probability thresholds <0.38 were all correctly classified as no pain (n = 9), whereas those with probabilities >0.65 correctly classified pain in 12 out of 14 discs (Fig 3). Building on a novel MRI method for non-invasive quantification of disc deformation [5], this study showed that disc deformation has the potential to phenotype/target discogenic pain in patients with nonspecific LBP. Such a biomarker could potentially guide surgeons in decision-making regarding therapy.
The study also moves the current research front forward by providing in vivo evidence of a close relationship between disc deformation and annular fissuring. While most discs with posterior fissures only displayed similar deformation as non-fissured discs, with compression posteriorly and expansion anteriorly, discs with fissures both anteriorly and posteriorly, and those with severe fissuring displayed large anterior deformation under load. While this deviant deformation pattern was clearly able to distinguish identify pain signaling discs, the results require further validation. This study supports the hypothesis that disc deformation, quantified with loading-based MRI, may be an image biomarker linked to disc-specific pain-signaling. Especially elevated anterior deformation associated with extensive fissuring, seems to be of value in identifying the pain-signaling discs. Such a precise biomarker of nonspecific LBP can lead to improved diagnostics, and potentially also better treatment outcomes for this large patient group.
Kerstin LAGERSTRAND (Gothenburg, Sweden), Hanna HEBELKA, Helena BRISBY, Christian WALDENBERG
11:00 - 12:30
#47790 - PG506 Low-cost, open-source, MRI-compatible grip force sensor for NMES-synchronised dynamic muscle MRI.
PG506 Low-cost, open-source, MRI-compatible grip force sensor for NMES-synchronised dynamic muscle MRI.
Synchronising dynamic MRI acquisitions with physical force measurements and neuromuscular electrical stimulation (NMES) enables standardised and normalised assessment of muscle activity [1] but requires an MR-compatible, e.g., safe and artefact-free setup with force sensors and NMES. While the NMES setup is generalised to dynamic MRI, the force sensor needs to be tailored to the respective muscle group under investigation. Building on a custom foot pedal sensor previously used for leg muscles [2], this study presents a further developed open-source, low-cost, MR-compatible grip force sensor for dynamic forearm muscle MRI.
Sensor design and setup: A handheld device was designed in FreeCAD (v0.21.2) and 3D printed (A1, Bambu Lab, Shenzhen, China) to house the force measurement components. The grip force measurement is implemented with two 50 kg (490 N) aluminium beam load cells in a parallel Wheatstone bridge configuration. An Ethernet cable connects the load cells to the electronics outside the scanner room. Furthermore, the subject is in head-first (superman) position so that the cable length within the scanner bore is minimised.
A custom low-pass filter protects the microcontroller (Arduino Uno [3]) from MRI interference. The microcontroller, placed outside the scanner room, streams force data to a PC via USB. A custom Python program (v3.13.0) logs the data.
The setup is extended by placing an (optional) NMES device (EM 49, Beurer GmbH) outside the scanner room. Detailed building instructions are available at [4], [5]. Fig. 1 shows a schematic of the electronics and assembly.
Testing the potential interaction between the grip force sensor and the MR scanner: The sensor was calibrated and tested for accuracy both inside and outside the MRI by systematic placement of known weights ranging from 0.1 to 100 kg outside the scanner room and up to 10 kg MRI-compatible weights inside the MRI while a GRE sequence was running.
To evaluate the MR compatibility of the force sensor, water phantom measurements and in-vivo measurements were conducted with a 3 T whole-body MRI scanner (MAGNETOM Prisma, Siemens Healthineers) and a 18-channel flex coil for signal acquisition.
For the phantom-based measurements, a dual echo, gradient-recalled echo (GRE) sequence was used to calculate B0 maps and SNR with and without the setup active. For both scenarios, a Siemens RF noise service sequence was used to acquire RF noise spectra. Following NEMA [6] standards, the SNR was calculated from ten separate acquisitions of signal and pure noise, which were subsequently used for a t-test.
To assess the functionality of the whole setup, a conventional 2D phase contrast sequence was acquired sagittally while triggering with NMES in five healthy subjects. The force sensor demonstrated good linearity with an R² value of 0.998 / 0.999 inside and outside of the MRI (Fig. 2). Minor B0 inhomogeneities were observed with the installed setup and active sensor compared to the baseline. SNR decreased by 3.7% (p < 10⁻⁴) from 232 ± 5 to 223 ± 10 with the sensor active. Mean and max RF noise remained similar with and without the device (Fig. 3).
The setup reliably recorded the force profile of each voluntary contraction during dynamic MRI. The magnitude of the measured force varied intersubjectively. Furthermore, in subjects one, three and five, a gradual decrease in contraction force is observed (Fig. 4). With consistent linearity and accuracy, the sensor suits dynamic MRI tasks requiring a broad range from 20% of normal grip force ranges of 11 kg/ 110 N evoked by NMES to a maximum voluntary grip force of up to 83 kg/ 814 N [7], [8].
The low-pass RF filter is essential for a reliable force recording. Without the filter installed, the microcontroller experienced random resets. Furthermore, uncontrolled motion and poor cable management introduced falsely recorded force values, including negative force values and RF-induced offsets. This can be addressed by proper wrist stabilisation with vacuum paddings and minimising the length of the Ethernet cable within the scanner bore (direct RF field).
The utilisation of the grip force sensor caused a 3.7% drop in SNR, which is acceptable for grip force measurements but may be problematic for lower SNR applications like non-proton imaging or spectroscopy. The associated B0 variations were minor compared to B0 inhomogeneities induced by differences in susceptibility of anatomical structures in various regions of the body [9], [10]. In this paper, we presented an open-source implementation of a grip force sensor using commercially available components not specifically designed for the application in an MRI environment. The device demonstrated robust functionality and a minimal penalty in terms of output image quality. Therefore, it is feasible to utilise the grip force sensor with optional NMES in dynamic muscle MRI to characterise forearm muscle activity in subjects.
Sabine Melanie RÄUBER (Basel, Switzerland), Marta Brigid MAGGIONI, Francesco SANTINI
11:00 - 12:30
#47821 - PG507 Low-field knee MRI in the clinical setting: a comparative study of a 72 mT with a 3 T scanner.
PG507 Low-field knee MRI in the clinical setting: a comparative study of a 72 mT with a 3 T scanner.
Low-field magnetic resonance imaging (LF-MRI) systems stand out for their portability and cost-effectiveness, albeit at the expense of reduced signal-to-noise ratio (SNR), or resolution, when compared to high-field systems. Despite growing interest in LF-MRI development, its diagnostic utility remains largely unexplored. In this study, we present an on-going investigation of the clinical potential of LF-MRI for musculoskeletal imaging of patients with knee lesions. To this end, we are employing a 72 mT system called Physio I [1]. All patients are also being scanned in a Philips ACHIEVA 3T clinical scanner [2]. The comparison between the paired images will allow us to determine the diagnostic potential of our Point of Care(PoC) system. The study is performed at La Fe Hospital in Valencia.
A cohort of at least 80 patients with undiagnosed knee injuries is presently undergoing MRI scans on both systems. These images will be compared to evaluate the diagnostic capacity of our LF system.
Our LF protocol for Physio I is optimized for clinical diagnosis, aligning with the sequences of the 3T system in La Fe while ensuring sufficient resolution, SNR, and total duration. It includes four sequences: a T1-weighted Rapid Acquisition with Relaxation Enhancement (RARE) sagittal scan, a T2-weighted RARE sagittal scan, and both sagittal and coronal Short Tau Inversion Recovery (STIR) sequences, for a total duration of approximately 40 minutes (Fig.1). These sequences will be evaluated by at least one radiologist without prior knowledge of the injuries present, and compared with the diagnosis from the 3 T acquisitions.
Technically, several key improvements have been implemented in our LF system since a prior study on healthy volunteers [3]: the x-gradient coil was extended to increase the axial field-of-view (FOV), the RF coil elements were lengthened, and sequence parameters optimized for better contrast and SNR. An in-depth eddy-current analysis led to mechanical redesigns, reducing aluminium parts and suppressing related artefacts. For improved patient comfort, we added bed-positioning rails and a screen for a better overall experience. At the time of writing this abstract, we have successfully installed our system, Physio I, in La Fe Hospital (Fig.2a). Once there, we optimized the positioning and configuration of our system to minimize electromagnetic noise, and trained the radiology technicians in patient handling for Physio I. So far, we have scanned 6 out of the intended 80. These initial images show promise in distinguishing anatomical structures and musculoskeletal pathologies (Fig.2). We observe certain distortions and artifacts due to the inhomogeneity of B0 and non-linearities of the gradient fields. However, these can be corrected using both Single-Point Double-Shot methods (SPDS, [4]), and geometrical co-registration algorithms based on Elastix [5].
As we are still in the data acquisition phase, no post-processing has been applied, and diagnostic evaluations are pending. At the moment, we are building the database by storing the acquired knee paired images, following a common folder convention between our data and the data acquired in Philips ACHIEVA. This paired 72 mT and 3 T dataset will then be used as training data for machine learning applications, including domain translation networks, among others, aiming to improve the perceived resolution and diagnostic value of low-field images. Additionally, it will be used to evaluate the diagnostic capability of our LF images with respect to the high-field images, following an evaluation performed by radiologists from La Fe hospital. This is the first comparative clinical study evaluating the diagnostic performance of a low-field scanner for musculoskeletal knee applications with patients presenting real injuries. Although the project is still in the data-acquisition phase, the preliminary results obtained with our 72 mT Physio I scanner show potential to distinguish important anatomical features of the knee, as well as different injuries. As we scan patients, we are building the first muskuloskeletal knee database of paired images from low to high field strength. After the data acquisition phase, we will use said database to provide insight into the clinical viability of LF systems, as well as the potential of AI-based methods to enhance the contrast and increase the quality of LF images.
Marina FERNÁNDEZ-GARCÍA, Teresa GUALLART-NAVAL, Amadeo TEN ESTEVE, Sonia GINÉS CÁRDENAS, Jose BORREGUERO PLAZA, Lorena VEGA CID, Luiz GUILHERME DE CASTRO SANTOS, Lucas SWISTUNOW, Eduardo PALLÁS LODEIRO, Jesús CONEJERO RODRIGUEZ, Jose M. ALGARÍN, Fernando GALVE CONDE, Luis MARTÍ-BONMATÍ, Joseba ALONSO OTAMENDI, Elisa CASTANON GARCIA-ROVES (Valencia, Spain)
11:00 - 12:30
#46625 - PG508 Fast Field-Cycling as a Tool for Investigating Fibrin Clot Microstructure.
PG508 Fast Field-Cycling as a Tool for Investigating Fibrin Clot Microstructure.
Fast-field cycling NMR relaxometry (FFC-NMR) measures T1 over a wide range of low magnetic field strengths (Figure 1), typically between 25 µT - 200 mT, to produce R1 (1/T1) NMR dispersion profiles (NMRD) that graphically show T1 relaxation changes with the magnetic field strength. While nuclear magnetic resonance dispersion (NMRD) profiles acquired using FFC-NMR reflect molecular dynamics of water occurring on different timescales (ms to µs), the biological meaning and physiological importance is unclear.
We investigated whether and to what extent biologically relevant characteristics of protein gels could be quantified from NMRD profiles. We used a fibrin clot as a model as it is a widely studied protein system. In fibrin clots, water molecules demonstrate a spectrum of molecular dynamics, inclusive of adsorption and desorption motion [1,2]. Relaxation of such motion is thought to be mediated via reorientation mediated by translational displacements (RMTD) [1,2], an expression that characterises relaxation of adsorption and desorption motion using the diffusion constant of water molecules interacting with proteins, fractal dimension (integer characterising geometric complexity), and resonance frequency.
It remains uncertain if interactions between water molecules and the fibrin fibres can be detected using FFC-NMR, and whether such adsorption and desorption motion can be reliably described by RMTD. Moreover, the dominant timescale where water molecules continuously transfer between bulk and fibrin fibre surfaces is unknown.
Therefore, this study aimed to model NMRD profiles of fibrin clots using the RMTD expression to retrieve the fractal dimension and quantify the microstructure of fibrin network.
To determine if RMTD was dominant in the NMRD profiles of fibrin clots, we compared the fractal dimension obtained using the RMTD expression with the one obtained from confocal microscopy images. Fibrin clots comprised of various concentrations of thrombin (0.01–1U/mL) and fibrinogen (1–3mg/mL) were measured using FFC-NMR relaxometry at 25 – 40°C across 15 different magnetic fields. Measurements were acquired using both pre- and non-polarised multi-exponential sequences with a change at 4 MHz succeeding polarisation (Figure 1).
Confocal microscopy of fibrin clot with fluorescent-labelled fibrinogen were then generated in chamber slides and imaged to validate information from RMTD expression. Gold standard image analysis algorithms were used to obtain reference fibrin clot fractal dimensions. Confocal images (Figure 2) validated structural changes of fibrin clots in accordance with the concentration of fibrinogen or thrombin used for clot formation. High concentrations of thrombin and fibrinogen produced dense clots composed of highly branched and thin fibres, whereas lower concentrations produce course networks of unbranched and thick fibres. The differing fractal dimensions between all fibrin clots retrieved using confocal microscopy image algorithms (Figure 3) further quantitatively validated the structural difference.
Moreover, the microstructural differences between fibrin clots were measurable using FFC-NMR, as distinct differences in NMRD profiles were evident (inclusive of inflection points, gradients, and offsets). We identified three different molecular motions dominating (inflection points of NMRD profiles) between 25 µT – 200 mT: regime 1 (200 mT and 10 mT), regime 2 (10 mT and 100 µT), and regime 3 (100 µT and 25 µT).
Structural information of fibrin clots from FFC-NMR between 10 mT – 100 µT was equivalent to confocal microscopy (p > 0.05). The fractal dimensions for all fibrin clots found using RMTD expression compared to confocal microscopy between these magnetic fields were 1.70 (±0.1) and 1.68 (±0.09), respectively (Figure 3). This work shows the relaxation of fibrin clots between 10 mT – 100 µT is likely dominated by rapid adsorption and desorption of water molecules between the bulk and surface of fibrin fibres, RMTD. The general structure of fibrin clots can be characterised in this region by modelling NMRD profiles with RMTD expression. Outside this field range, other molecular motions dominate and may be better studied with alternative models. Determination of fibrin clot microstructure is crucial in predicting the likelihood of rupture, embolization or response to antithrombotic management. Correctly identifying the motional water regimes over the frequency spectrum will ultimately contribute to assessing the potential future use of low-field imaging of blood clots, as well as other pathological protein networks.
Madeleine RHODES (Aberdeen, United Kingdom), Nicola MUTCH, Lionel BROCHE
11:00 - 12:30
#46975 - PG509 MR-based characterization of Giant Unilamellar Vesicles as synthetic cell phantoms.
PG509 MR-based characterization of Giant Unilamellar Vesicles as synthetic cell phantoms.
The assessment of cell size and cell packing via diffusion-weighted MRI (dMRI) have proven useful to monitor tumor progression and anti-tumor treatment efficacy.[1,2] The development and refinement of dMRI techniques and biophysical diffusion-signal models has thus far mainly been performed on aqueous suspensions of (yeast) cells or polystyrene latex beads.[3,4] While cell suspensions best replicate cellular structures, their biophysical and chemical properties are difficult to control over time and between batches. Furthermore, preparations of individual samples can take several days to weeks. Bead-filled phantoms are faster to produce with controlled diffusion dimensions but the bead material does not replicate biological membranes in elasticity and permeability. To address these issues, we propose giant unilamellar vesicles (GUVs) as synthetic cell phantoms.
GUVs were prepared in an aqueous solution via the hydration method. Their membranes were composed of 10 mg/mL phosphatidylcholine (POPC) and 1 mol% biotin. Different size classes were achieved after gravity-filtration through a 10 µm mesh. GUV samples were condensed into visible patches by adding 5-10 µg/mL streptavidin to the suspension and brief centrifugation. The supernatant of a subset of unfiltered, H2O-filled GUVs was replaced with >99%, 50% or 25% D2O to determine the impact of the extravesicular compartment on dMRI measurements. Patches in suspension were imaged in a Bruker BioSpec 94/20 MR scanner using a 2-element cryoprobe with pulsed and oscillating gradient spin echo sequences with segmented EPI readout (PGSE and OGSE, respectively, fmax = 200 Hz, bmax = 2000 mm²/s). GUV radii and packing were calculated using the IMPULSED model.[5] IMPULSED-derived radii were compared to radii obtained from fluorescence microscopy of the same samples. Since larger GUVs contribute more to the MR-derived radii, microscopy radii were volume-weighted to allow for meaningful comparisons (effective radii reff).[6] To confirm the samples’ chemical composition, we determined relaxation times T1 and T2 in the GUV patches and recorded localized 1H NMR spectra (STEAM; 3×1×3 mm³ voxel, 512 averages, TE/TR = 3/4000 ms, VAPOR water suppression). Aggregated GUV patches were always visible in dMRI as hyperintense areas, characterized by reduced T2 and unchanged T1, compared to the standard medium (fig. 1). IMPULSED-derived radii and microscopy radii for filtered samples were smaller than for unfiltered samples (fig. 2A, B). With respect to IMPULSED radii, this difference was significantly larger for scans using sine-modulated OGSE dMRI sequences compared to scans using cosine-modulated OGSE. IMPULSED radii did not differ significantly from microscopy radii. While the slope of the linear regression between microscopy radii and sine radii across filtered and unfiltered samples was only 0.47, the regression slope was 0.90 for cosine radii, albeit at a wider confidence interval (fig. 2C, D). Localized 1H NMR spectra revealed three peaks upfield of the water resonance frequency (fig. 3).
H2O-filled GUVs suspended in >99% D2O were hyperintense even in T2-weighted spin echo sequences. Despite very low overall signal in the >99% D2O samples, GUVs were still visible in high b-value and high frequency dMRI. T2 was reduced by 40% at >99% D2O but ADC was slightly elevated, compared to 50% and 25 % D2O suspensions. Low signal intensities corresponded to a larger spread of IMPULSED radii in the GUV patch at >99% D2O (fig. 4). In all cases, radii outside of the patch converged on the fit limits (0 µm and 30 µm; fig. 1 & 4). Using multiple MR modalities, we have characterized key physicochemical parameters of the GUV system. Compared to conventional phantoms for diffusion-based cell size measurements in MRI, GUVs can be synthesized within hours. Streptavidin-biotin-assisted packing is sufficient to form macroscopic patches with reduced diffusivities and T2. Here, we confirmed that the IMPULSED model is suitable to detect size differences of GUVs following gravity-driven filtration and that IMPULSED radii are in good agreement with radii determined from microscopy. 1H spectra of cross-linked GUVs in suspensions show peaks for the three main constituents of POPC, however, peaks are relatively broad.[7]
Replacing the extravesicular medium with D2O reduces overall signal, however, IMPULSED can still be successfully applied after sufficient averaging. Doping the intravesicular or extravesicular medium with signal suppressing D2O presents a relatively straight-forward route to disentangle contributions of different compartments to diffusion signals. This may open the possibility for calibrating signal models under well-defined in situ conditions. GUVs are a promising multicompartment tool for dMRI-based cell size measurements that can be produced within hours at desired size ranges, packing and chemical composition.
Bastian MAUS (Münster, Germany), Daniele DI IORIO, Robert VORNHUSEN, Seraphine V. WEGNER, Cornelius FABER
|
Poster hall |
12:30 |
LUNCH BREAK
|
13:30 |
"Saturday 11 October"
Added to your list of favorites
Deleted from your list of favorites
B34
13:30 - 15:30
CAMERA
13:30 - 13:40
Welcome remark.
Udunna ANAZODO
13:40 - 14:00
Real-world implementation of open source MRI designs.
Guillermo SAHONERO ALVAREZ (Keynote Speaker, Santiago de Chile, Chile)
14:00 - 14:20
Multisite construction of open source preclinicla scanners.
Maureen NAYABERE (Keynote Speaker, Chile)
14:20 - 14:40
Engineering low-field MRI for resource constrained settings.
Zhiyong ZHANG
14:40 - 15:00
Perfusion MRI in resource constrained settings.
Danny Jj WANG (Keynote Speaker, USA)
15:00 - 15:30
Interactive discussion : implementation of open source MRI in RCS.
Martina FERNANDEZ GARCIA (Keynote Speaker, Spain)
|
Espace Vieux-Port |
|
"Saturday 11 October"
Added to your list of favorites
Deleted from your list of favorites
D34
13:30 - 15:00
ET2-3 - Low Field MRI
13:30 - 15:00
Cardiac at low field.
Amedeo CHIRIBIRI (Keynote Speaker, United Kingdom)
13:30 - 15:00
Low field MRI - How? - Radiographer's perspective on sequence optimisation at LF.
Anastassia KOROLENKO
13:30 - 15:00
Low field MRI - Why? - Reasons forand against low field MRI.
Joan C (Kai) VILANOVA (Chief) (Keynote Speaker, Girona/ES, Spain)
13:30 - 15:00
MSK at low field.
Christian BREIT (Keynote Speaker, Switzerland)
13:30 - 15:00
Neonatal at low field.
13:30 - 15:00
Neuro at low field.
Edmond KNOPP (Chief Medical Officer) (Keynote Speaker, White Plains, USA)
|
Salle 120 |
|
"Saturday 11 October"
Added to your list of favorites
Deleted from your list of favorites
G34
13:30 - 15:00
Poster 7
FT2 - Aging Across the Lifespan | FT2 - Neurodegenerative Diseases | FT2 - Multiple Sclerosis | FT1 - Spine and body | FT1 - Brain and neuroscience
13:30 - 15:00
#47575 - PG510 Evaluation of the therapeutic effect of stem cells on an animal model of neonatal brain injury by advanced diffusion MRI.
PG510 Evaluation of the therapeutic effect of stem cells on an animal model of neonatal brain injury by advanced diffusion MRI.
Preterm infants represent the largest patient cohort in pediatrics. Despite increased survival rates due to advances in neonatal intensive care, the risk of long-term complications such as encephalopathy of prematurity (EoP) remains high. Inflammation as well as high oxygen concentrations (hyperoxia) are major risk factors for premature birth and preterm birth related brain injury. Hyperoxia and inflammation induce perinatal brain injury, affecting white and gray matter structures differently. Currently, there is no causal therapy available. Mesenchymal stem cells have shown promising neuroregenerative potential in preclinical models. The aim of this work was twofold, 1st, to evaluate brain alterations in our model of neonatal brain injury and 2nd, to assess the potential neuroprotective effect of human umbilical cord-derived mesenchymal stem cells (HMSCs) on it.
Pregnant rats received an intraperitoneal (i.p.) injection of lipopolysaccharide (LPS, 100 µg/kg) or sodium chloride (NaCl) at embryonic day 20. Newborn pups were then exposed to either normoxia (21% O₂) or hyperoxia (80% O₂) from postnatal day 3 (P3) to P5. Human MSCs (50 × 10⁶ cells/kg) were administered intranasally at P5.3 groups were finally assessed: Sham (NaCl+normoxia), LPS (LPS+Hyperoxia) and HMSC (LPS+Hyperoxia+MSCs therapy). Corresponding to term-equivalent age in humans, rat brains were collected at P11. Myelination and neuroinflammatory responses were assessed using immunohistochemistry and western blot. To evaluate long-term effects, motor-cognitive behavioural tests were performed during adolescence and adulthood (P40). Following completion of behavioural testing, brains were examined ex-vivo using advanced diffusion MRI to detect microstructural alterations. Ex-vivo MRI experiments were performed with a 2.5 cm diameter birdcage coil, on an 9.4T/31cm actively shielded horizontal-bore magnet (Magnex Scientific, Yarnton, UK), B-GA12S HP shielded gradient set (114mm ID, 660 mT/m peak strength and 4570 T/m/s slew rate, Bruker BioSpin, Ettlingen, Germany) interfaced to a Bruker BioSpec console with AVANCE NEO electronics running ParaVision 360 v3.5. A multi-b-value shell protocol was acquired using a spin-echo sequence (FOV = 23 × 18 mm2, matrix size = 128 × 92, 18 slices of 0.6 mm, 3 averages with TE/TR = 22/1800ms). 82 DWI were acquired, 6 b0 images and 76 separated in 3 shells (noncollinear and uniformly distributed in each shell) with number of directions/b-value in s/mm2: 16/1500, 30/3000 and 30/6000. DTI metrics including Axial diffusivity (AD), Radial diffusivity (RD), and Fractional anisotropy (FA) were derived from the tensor. We also calculated neurite orientation dispersion and density imaging (NODDI) metrics using AMICO, including Neurite density index (NDI), Orientation dispersion index (ODI), and Free water fraction (FWF). These parameters were calculated for 5 Ipsilateral ROIs for each animal (Figure): corpus callosum (CC), cingulum (Cg), external capsule (EC), junction form corpus callosum to external capsule (CC_EC), primary motor cortex (M1Cx) and primary somatosensory cortex (S1Cx). Data were averaged for the coronal planes and statistically tested between groups using one-way ANOVA tests with Tukey’s post-hoc. Double-hit model showed decreased fibre length and reduced branching points of myelin basic protein (MBP)-positive fibres reversed by MSC treatment Indeed, an increased microglial activation was observed in the LPS group, indicated by increased Iba1 and CD68 double-positive cells, which was markedly reduced after MSC application. Adolescent and young adult animals exposed to the double-hit showed memory impairments, associated with altered white matter microstructure as detected by DTI (Figure). Mainly, in the CG and CC_EC regions, increased RD and ODI as well as decreased FA and NDI were observed in the LPS group compared to both Sham and HMSC groups. We demonstrated that the combination of prenatal inflammation and postnatal hyperoxia (double-hit) significantly reduces brain volume, likely due to impaired myelination. Both behavioural and structural alterations were alleviated by neonatal MSC treatment as suggested also by MRI results. Our findings suggest that intranasal MSC therapy mitigates both early and long-term structural and functional brain impairments in a rat model of EoP. These findings support the translational potential of MSC therapy for neonatal brain injury in preterm infants.
This project has received funding from the European Union’s Horizon 2020 Research and Innovation programme under Grant Agreement No 874721.
Yohan VAN DE LOOIJ (Geneva, Switzerland), Meray SERDAR, Ursula FELDERHOFF-MÜSER, Ivo BENDIX
13:30 - 15:00
#47748 - PG511 ¹H MRS at 3T reveals impact of childhood adversity on age-related neurometabolic changes in the hippocampus and posterior cingulate cortex.
PG511 ¹H MRS at 3T reveals impact of childhood adversity on age-related neurometabolic changes in the hippocampus and posterior cingulate cortex.
Maintaining metabolic balance in the human brain is vital for cognitive and neurological function, yet this equilibrium shifts with advancing age [1], altered neurotransmission, and the onset of neuroinflammatory processes [2]. These metabolic alterations may also be affected by early environmental influences, such as early-life stress (ELS) that includes experiences of neglect or abuse. ELS is recognized for its detrimental effects on development and long-term brain health [3] and can also alter both, cognition and emotional processing [4]. Understanding the interplay between ELS and age-related neurometabolic changes could offer new insights into individual trajectories of brain health across the lifespan. 1H MR spectroscopy (MRS) provides a non-invasive means to assess neurochemical processes in vivo [5] and is well-suited to identifying biomarkers associated with ELS. The hippocampus (HC) and posterior cingulate cortex (PCC) were selected – two regions central to cognition and emotion, and known to be sensitive to early adversity. The PCC, a metabolically active hub of the default mode network [6], is associated with cognitive impairment [7], while the HC is susceptible to stress-induced dysregulation and structural damage [8]. Despite the suggested link between age, brain metabolism changes, and neuropathology progression in later life, the role of ELS in this context remains underexplored. Thus, this study examined whether ELS interacts with age to influence neurometabolic profiles in adulthood, as measured by 1H MRS. Preliminary results from the female cohort have been recently reported [9].
135 adults (79f, aged 30–60 yrs), with varying severity of ELS exposure, were recruited for structured interviews and questionnaires and MRS acquisitions.
ELS Assessment: Exposure to ELS (occurring prior to the onset of puberty) was ascertained using the Childhood Trauma Questionnaire (CTQ) [10].The total CTQ score that assesses five different categories of childhood maltreatment was used, since with its high granularity subtle variations in trauma severity can be captured.
MRS: Scans were performed on a 3T PrismaFit system (Siemens Healthineers, Erlangen, Germany) using a 64 channel radiofrequency (RF) coil. High-resolution T1-weighted MPRAGE images were used for voxel placement. For 1H MRS, localized RF calibration was performed, and first- and second-order shims were adjusted using FAST(EST)MAP [11]. Single volume spectra were acquired from the HC and PCC using the semi-LASER technique [12] (TR/TE = 3000/23 ms, spectral width = 2000 Hz, VOIHC = 10x12x35 mm3, number of averages (NA)HC = 256, VOIPCC = 20x20x20 mm3, NAPCC = 128). MRS data were pre-processed utilizing the FID-A toolkit [13]. Metabolites of resulting spectra were quantified using LCModel [14] with a simulated basis set.
Statistics: Generalized Additive Models (GAMs) [15] were applied for statistical analyses. Using tensor product smoothing, GAMs were fitted to the metabolite data to model the interaction between age and CTQ scores – both as continuous predictors – using the Restricted Maximum Likelihood method. All statistical analyses were run in R Project for Statistical Computing (R Core Team) with the significance level set at p<0.05. Shimming resulted in water linewidths of 8.1 ± 0.8 Hz (HC) and 6.0 ± 0.4 Hz (PCC), respectively. Cases with poor shim in the HC (LWH2O ≥ 10 Hz) or strong artifacts were excluded, such that NHC = 117 and NPCC = 133. The overall high spectral quality (Fig. 1) allowed quantification of neurochemical profiles in both regions including several metabolites (Fig. 2). Using GAMs, highly non-linear interactions between CTQ score, age, and metabolite levels were observed. Significant effects were found for glutamate (Glu) (p = 0.02), N-acetylaspartate (NAA) (p = 0.01), and myo-inositol (Ins) (p < 0.001) in the HC (Fig. 3), and for total choline (tCho) (p = 0.004), and Ins (p < 0.001) in the PCC (Fig. 4). This study examined whether age interacts with ELS severity to affect neurometabolic profiles in adulthood. Key findings revealed that GluHC, NAAHC, and tChoPCC levels were highest in younger adults with high maltreatment severity. Notably, choline—a marker of membrane turnover—was elevated, potentially reflecting myelination breakdown [16]. Ins showed a bimodal pattern across regions, echoing prior evidence of non-linear glial responses in neuroinflammation and degeneration [17,18]. NAA levels, while typically reduced in neurodegeneration, showed compensatory increases, possibly reflecting early adaptive responses to stress [19]. Altered Glu in the HC suggests early astrocytic engagement in managing excitotoxic stress [20]. Together, these patterns indicate subtle, early metabolic shifts linked to ELS, particularly in the context of aging, which may precede structural or cognitive decline. Exposure to ELS can be linked to age-related alterations in specific brain metabolite concentrations in adults.
Ralf MEKLE (Berlin, Germany), Lara FLECK, Martin BAUER, Dinesh K. DEELCHAND, Claudia BUSS, Sonja ENTRINGER, Jochen B. FIEBACH, Matthias ENDRES, Christine HEIM
13:30 - 15:00
#46034 - PG512 Exercise-induced changes in resting state functional brain activity in children with concussion.
PG512 Exercise-induced changes in resting state functional brain activity in children with concussion.
Over the past decade, the approach to managing concussion has evolved. Rest was traditionally considered the most effective treatment. However, recent research suggests that exercise may be a key component of effective concussion management1,2. Sub-maximal aerobic exercise has been shown to reduce symptoms and improve outcomes1, but there remains limited understanding of how this impacts functional brain activity in pediatric patients. Resting-state functional MRI (rsfMRI) is a valuable tool for assessing these changes, as it provides insight into brain connectivity patterns post-injury3. Data in adults suggests that there is a differential rsfMRI response to exercise in those with concussion compared to healthy controls4. Our study aims to explore the effects of sub-maximal aerobic exercise on resting-state functional brain activity in children with concussion.
A prospective cohort study was conducted with children experiencing their first concussion within four weeks of injury. Participants were age- and sex-matched with healthy controls and it was verified that all were without any prior neurological conditions. Two study visits were completed. In the first visit, sub-maximal aerobic exercise was
performed using the Buffalo Concussion Treadmill Test5, to determine each individual subject’s symptom-limited exercise threshold. During the second visit, imaging was conducted using a 3-Tesla GE Discovery MR750 scanner with a 32-channel phased array head coil (General Electric Healthcare, Milwaukee, WI). A 3D T1-weighted anatomical MRI (1mm isotropic IR-prepped fSPGR,TE/TR/TI/flip angle = 7.5ms/2.1ms/450ms/12o, 1mm slice thickness, 256x256, 25.6cm FOV) and resting-state fMRI (gradient echo EPI, TE/TR/flip angle = 35ms/2000ms/90degrees, 64x64matrix, 4mm thick, 24cm FOV, 5 discarded volumes, 240 volumes= 8:10min) scans were acquired, with preprocessing in CONN, involving registration and normalization, slice-timing correction, outlier and artifact detection, and spatial smoothing (8mm FWHM). More specifically, during this secondvisit, a baseline resting-state fMRI was obtained, followed by 10 minutes of treadmill exercise and (within 2-minutes of completing exercise and transit to the MR room) a post-exerciseresting-state fMRI to allow for an understanding of within-subject and between-group pre-post exercise changes in rsfMRI. The study was approved by our local research ethics board. Preliminary data from 9 participants included five children with concussion (3 males, average age 14.8±2.1, with an average symptom score of ) and four healthy controls (2males, average age 14.2±2.3). Seed-based analysis in the posterior cingulate cortex (PCC) revealed no significant pre-post exercise changes in connectivity for healthy controls. Children with concussion, however, exhibited hypoconnectivity in regions including the inferior parietal lobule and post-central gyrus after exercise. Group-level
analyses indicated more pronounced hypoconnectivity in the concussion group compared to controls. Discussion: Based on these findings we suggest a differential response in rsfMRI following exercise between children with concussion and healthy controls. Children with concussion demonstrated notable hypoconnectivity post-exercise, whereas healthy participants did not show similar changes. These results are consistent with studies in adults with mild traumatic brain injury3, suggesting lasting impacts of concussion on brain connectivity. Understanding these exercise-induced changes may inform targeted interventions aimed at both symptom management and enhancing brain health during concussion recovery. Our results are preliminary with further data analyses pending. Initially, these results converge with prior data on adults, with the concussion group demonstrating pre-post exercise hypoconnectivity not observed in controls. The impact of these changes on other clinical outcomes remains to be explored. Our preliminary findings may significantly impact rehabilitation strategies for pediatric concussion. If confirmed in a larger sample (recruitment is ongoing), we suggest that while
sub-maximal aerobic exercise aids in symptom reduction, it may also alter brain connectivity in ways that need further investigation. These insights could support the development of personalized exercise prescriptions to optimize brain health and functional recovery in children post-concussion.
Bhanu SHARMA (Hamilton, Canada), Eric KOELINK, Carol DEMATTEO, Brian TIMMONS, Michael NOSEWORTHY
13:30 - 15:00
#47874 - PG513 White Matter Alterations in Interhemispheric Tracts Predict Age-Related Decline in Proprioception.
PG513 White Matter Alterations in Interhemispheric Tracts Predict Age-Related Decline in Proprioception.
Proprioceptive deficits have been reported to emerge in normal aging after the age of 65, with a specific impairment in the ability to discriminate movement velocity [1]. A previous fMRI study showed reduced lateralization of primary sensorimotor activations in older adults during unilateral proprioceptive stimulation when compared to young adults [2]. Moreover, this reduction in lateralized activity patterns in older adults was found to correlate with proprioceptive decline [1,2]. Although numerous studies have described microstructural alterations in the brain associated with aging [3], the potential link between these structural changes and proprioceptive decline remains to be established.
In the present study, we explored the extent to which alterations in interhemispheric structural connectivity between the two sensorimotor cortices might account for the loss of functional hemispheric laterality observed in normal aging. To this end, we examined the relationship between age-related White Matter (WM) alterations and proprioceptive decline by combining diffusion-weighted imaging (DWI), task-related fMRI activity, and psychophysical measures obtained during proprioceptively induced hand illusions. The analysis specifically focused on the integrity of the Corpus Callosum (CC), which has been previously associated with inefficient interhemispheric communication [3], and on the interhemispheric fiber bundles connecting the primary somatosensory and motor areas.
Twenty young (20-28 years) and twenty older (65-75 years) healthy right-handed adults participated in the study. Psychophysical performance was assessed using the just noticeable difference (JND) in hand movement illusions, while functional lateralization was evaluated through the interhemispheric difference (IHD) index [2]. WM microstructure was analyzed using diffusion tensor imaging (DTI) and constrained spherical deconvolution (CSD), with a specific focus on the corpus callosum. In particular, a tractography analysis was conducted on the motor (CC4) and somatosensory (CC5) interhemispheric tracts. Quantification of FA values along the reconstructed tracts was performed using TractSeg [4], which segmented each bundle into 100 segments.
To further investigate the relationship between WM integrity and functional outcomes, an unsupervised machine learning approach was applied using K-means clustering. Prior to clustering, dimensionality reduction was carried out using a backward Sequential Feature Selection (SFS) method, enabling the identification of the five most informative structural features across the 100 tract segments for each callosal tract (CC4 and CC5). Whole-brain voxel-wise analyses (TBSS) [5] and region-of-interest (ROI) tractography confirmed that aging is associated with reduced fractional anisotropy (FA) and axial diffusivity (AD), alongside increased mean diffusivity (MD) and radial diffusivity (RD) within sensorimotor callosal tracts.
Cluster-based analysis performed on the five selected features for each fiber bundle revealed that the somatosensory tract (CC5) enabled a more robust and functionally meaningful classification of participants compared to the motor tract (CC4). Specifically, CC5-based clusters showed strong alignment with participants’ chronological age groups and demonstrated higher predictive accuracy for proprioceptive impairments (JND) and reductions in interhemispheric lateralization (IHD). These predictions were further validated through cross-validation procedures, supporting the reliability of the clustering approach. The present results confirm structural brain alterations associated with aging, characterized by reduced FA overlapping with increased RD, a pattern typically linked to myelin degradation [6]. This suggests that the reduced integrity of the corpus callosum and associated interhemispheric fibers in older adults may predominantly result from demyelination processes.
Age-related microstructural alterations within the CC5 somatosensory tract may explain the observed reduction in functional laterality, reflecting less specific neural recruitment during proprioceptive processing. Critically, alterations in the CC5 somatosensory tract, rather than in the CC4 motor tract, more strongly predicted age-related declines in proprioceptive acuity (JND) and reductions in sensorimotor lateralization (IHD). Unsupervised clustering based on DTI features from CC5 successfully discriminated between age groups and accurately predicted functional impairments, highlighting the critical role of somatosensory callosal fibers in maintaining proprioceptive function with aging. These findings support the hypothesis that age-related microstructural degeneration of somatosensory callosal fibers contributes to a decrease in the specificity of neural recruitment during proprioceptive processing, aligning with the dedifferentiation theory of aging [3].
Daniela PINZON (MARSEILLE), Nicolas CATZ, Caroline LANDELLE, Raphaëlle SCHLIENGER, Julien SEIN, Jean-Luc ANTON, Olivier FELICIAN, Anne KAVOUNOUDIAS
13:30 - 15:00
#47572 - PG514 Neuroprotection via stem cell therapy mitigates structural brain injury in preterm lambs: evaluation by advanced diffusion MRI.
PG514 Neuroprotection via stem cell therapy mitigates structural brain injury in preterm lambs: evaluation by advanced diffusion MRI.
Preterm birth is the leading cause of neonatal morbidity and mortality. Advances in perinatal medicine increased survival rate, consequently increasing the number of preterm infants at risk for life-long neurocognitive disabilities. The pathogenesis of preterm brain injury is multi-factorial, with chorioamnionitis and mechanical ventilation as important contributors. To increase mechanistic understanding and develop therapeutic strategies, we developed a long-term translational triple-hit ovine model for preterm brain injury. Sheep brains display similar gyrification patterns to human brains. Therefore, utilizing sheep models for studying perinatal brain injuries offers the advantage of lesions being closer in proximity to those found in human neonates. Stem cells are interesting candidate that could mediate neuroprotection / recovery following perinatal brain injury. The aim of this work was twofold, 1st, to evaluate early (6 weeks) and late (1 year) brain alterations in our model and 2nd, to assess the potential neuroprotective effect of human umbilical cord-derived mesenchymal stem cells (hMSCs) on it.
Preterm lambs were exposed to triple perinatal hits including intra-uterine lipopolysaccharides (LPS), preterm birth and mechanical ventilation (3HIT group, n=7). Animals were either sacrificed directly after preterm birth, after 72h of mechanical ventilation, or followed-up for 12 months. hMSCs were administered intravenously directly after preterm birth and/or intranasal after 6 weeks (3HIT+SC, n=10). A sham group in the same conditions as 3HIT with saline injection was also assessed (SHAM, n=10). During long-term follow-up MRI, gait analysis and behavioral tests were conducted and combined with post-mortem histology for microglia, myelin and oligodendrocytes. MRI experiments were performed in-vivo at 6 weeks and 1 year old on a clinical 3.0T Philips scanner with a 32 channels human adult head 1H radiofrequency coil. A multi-b-value shell protocol was acquired using a SE-EPI sequence (FOV = 120 × 120 × 57 mm3, matrix size = 80 × 80 × 38, 2 averages with TE/TR = 132/3439 ms). 131 diffusion weighted images were acquired, 3 b0 images and 128 separated in 2 shells with number of directions/b-value in s/mm2: 64/1000, 64/2000. Acquired data were fitted using DTI-TK [1] for the conventional DTI derived parameters as well as the spherical mean technique (SMT) model [2] for advanced diffusion. Several white matter tracts were assessed including internal capsule (IC), external capsule (EC) and cortico-spinal tract (CST). DTI derived parameters (Axial diffusivity (AD), Radial diffusivity (RD), Mean diffusivity (AD) and Fractional anisotropy (FA)) and SMT derived metrics (intra-axonal volume fraction (intra), intra-axonal diffusivity (diff), extra-axonal mean diffusivity (extramd) as well as extra-axonal transverse diffusivity (extratrans)) were averaged in the different regions assessed for all animals. A Mann-whitney test was used for significance between the groups (p<0.05). Diffusion maps are presented on figure 1. At 6 weeks (figure 2 – upper panel), among the significant changes between SHAM and 3HIT we found an increase in AD and RD, and a decrease in intra and diff, depending on the white matter region assessed. These changes were not observed between Sham and 3HIT+SC groups. At 1 year old, only few metrics were significantly changed between SHAM and 3HIT (figure 2 – lower panel). Microglial numbers were elevated following preterm birth and mechanical ventilation. Moderate histological alterations were observed. Structural brain injury correlated with aberrant behavior, assessed around adulthood. At 6 weeks, myelin impairments were evident in the evaluated white matter tracts, indicated by elevated diffusivity values (AD and/or RD) and reduced intra (neuronal density) and diff (intra-axonal diffusivity) in the injured group. Interestingly most of these changes were restored in the stem cells treated group. By the age of 1 year, these injuries appeared less severe, likely owing to defense mechanisms and well-established brain plasticity mechanisms characteristic of brain development. In this study we were able to show the phenotype of mild 3HIT induced lesion characterized by advanced diffusion MRI as well as the potential neuroprotective effect of hMSCs. Early intervention with hMSCs showed promising effects on white matter structure and neurocognitive functioning on the long-term, which is of potential clinical interest to improve quality of life for children born preterm.
This project has received funding from the European Union’s Horizon 2020 Research and Innovation programme under Grant Agreement No 874721.
Yohan VAN DE LOOIJ (Geneva, Switzerland), Valery VAN BRUGGEN, Rob WESTERLAKEN, Reint JELLEMA, Daan OPHELDERS, Tim WOLFS
13:30 - 15:00
#47144 - PG515 Age-related changes in structural and functional organization of the human spinal cord.
PG515 Age-related changes in structural and functional organization of the human spinal cord.
Aging is associated with changes in sensorimotor function1,2 and its neural correlates2,3,4, but most studies to date have focused on brain sensorimotor networks, often overlooking the spinal cord. In addition, age-related changes in spinal cord function have mainly been explored through electrophysiological approaches, which reveal reduced reflex loop responses5,6. However, this approach focuses on local circuitry, hence missing large-scale interactions between spinal segments. In the present study, we used spinal MRI7,8 in humans to investigate on a large-scale the age-related functional and structural changes at the cervical spinal cord level.
Sixty-seven healthy adults (36 females, 46.5 ± 16.8 yrs old) were included in this study. The experiment was approved by the local ethics committee and written informed consent was obtained. The acquisition protocol (Fig. 1) included spinal cord T2*w, magnetization transfer (MT) and multi-shell diffusion MRI sequences (DWI), as well as simultaneous brain/spinal cord T1w and functional imaging (3T Siemens Prisma scanner).
Preprocessing was performed using the Spinal Cord Toolbox (SCT)9 and included spinal cord segmentation, vertebral labeling of the T1w image, motion correction for functional and DWI data, co-registration for MT on/off images, and non-linear registration of the T1w image on the PAM50 template. Functional images were also denoised and filtered using a custom pipeline.
Diffusion metrics including fractional anisotropy (FA) and radial diffusivity (RD) were computed from the DWI images. Using the PAM50 spinal segmental atlas10 we extracted the average T2*w gray/white matter ratio, MT ratio, and diffusion metrics from each spinal level from C2 to C7 (Fig. 2a) and regressed them against age.
Spinal functional levels (C1–C7) were identified in a data-driven manner using the iCAPs framework8,11. A seed-based approach examined the age-related changes in functional connectivity (FC) between ventro-ventral (V-V), dorso-dorsal (D-D), ventro-dorsal ipsilateral horns (within) and ventro-dorsal contralateral horns networks (cross) across 28 seeds (4 horns x 7 levels). For each participant, mean denoised, deconvolved signal was extracted from the seeds, and Fisher-transformed correlation coefficients were used to generate individual FC matrices. The top 50% strongest correlations were retained for graph analysis with community structure identified via Louvain community algorithm12 (gamma = 1.8, 1000 partitions), followed by a consensus partitioning at the individual level. Linear regression analyses showed age-related decreases in all microstructural metrics (Fig. 2b, upper panel), including the GM/WM ratio, MT ratio, and changes in diffusivity measures such as decrease in FA and increase in RD (t=-3.07, p=0.003). Notably, these age-related changes were found significant across most spinal levels (Fig 2b, lower panel). The seven spinal cord levels were successfully identified using the iCAPs framework (Fig. 3a). The FC matrices revealed stronger intra-segment connectivity (within the same spinal level) compared to inter-segment connectivity (between different levels), with age-related increases in inter-segmental FC (Fig. 3b). To better interpret these FC changes, we applied graph theory analysis. Although modularity was not affected by aging (t=0.93, p=0.35), both local efficiency and centrality—reflecting the effectiveness and quantity of community connections—significantly decreased with aging (Fig. 2c). In addition, an increase in inter-communities connection strength was accompanied by a decrease in the ratio of intra- to inter-communities connection strength. This study provides first evidence of spinal microstructural changes13,14 using multimodal acquisition at different cervical spinal cord levels. Importantly, it also reveals age-related reorganization of functional connectivity. The relative increase in dorso-ventral FC, alongside inter-segmental FC likely reflects a reorganization of spinal networks with aging. While this may suggest compensatory mechanisms, graph-based analysis reveals a decrease in network segregation and FC specificity (reduced in weights ratio, local efficiency and centrality), alongside an increase in connectivity between the different spinal networks. These findings point to reduced synaptic efficiency and neural specificity, potentially driven by impaired inhibitory processes15,16. Further analyses that integrate structural and functional similarities, as well as brain-spinal cord functional connectivity, will be essential for a more comprehensive interpretation of these findings. This study provides the first opportunity to investigate age-related changes in the sensorimotor system, including the spinal cord. It offers valuable insights into sensorimotor integration across the lifespan and emphasizes the importance of distinguishing between healthy aging and disease effects in clinical research.
Caroline LANDELLE (Montreal, Canada), Nawal KINANY, Samuelle ST-ONGE, Ovidiu LUNGU, Benjamin DE LEENER, Véronique MARCHAND-PAUVERT, Julien DOYON
13:30 - 15:00
#47920 - PG516 NR2F1-associated neurodevelopmental disorder: computational neuroanatomy and cortical gyration.
PG516 NR2F1-associated neurodevelopmental disorder: computational neuroanatomy and cortical gyration.
The Bosch-Boonstra-Schaaf optic atrophy syndrome (BBSOAS) is a rare neurodevelopmental disease (< 1/1000000 birth resp.). It is classically characterized by developmental delay, intellectual deficiency and significant visual impairment due to optic nerve atrophy and hypoplasia or cerebral visual dysfunction. The spectrum of neurodevelopmental disorder is actually broader including autism spectrum disorder, learning disability without intellectual development disorder or epilepsy [1]. This syndrome is caused by loss-of-function variant of the NR2F1 gene, encoding a key factor in brain development. A French collaborative translational study of the BBSOAS has been initiated that already enabled to clarify the role of NR2F1 in cortical development and highlighted the existence of recurrent anomalies in the brain anatomy including abnormal cortical gyration such as dysgyria (excess and atypical organization of folds) [2]. However, these anomalies are still poorly described and understood. In this context, the present study proposes a first neuroanatomical insight in global and local cortical volumes and surfaces in the context of NR2F1 variant.
10 patients aged between 14 and 23 (mean age 18 y/o, sex ratio: 1) were imaged at NeuroSpin, CEA Paris-Saclay. 3D T1-weighted MRI were performed at 3T (Magnetom Prisma Fit, Siemens Healthcare) to obtain 0,9 mm isometric images. The dataset was completed with 3T 3D T1-weighted MRI of 77 typically developing controls from previous studies (12-18 y/o, mean age 16 y/o, sex ratio: 0,97), 40 of them with same site and sequence than the patients. The data were segmented using a pipeline combining Morphologist (BrainVISA [3]) and volBrain-AssemblyNet [4][5] into 6 cortical regions (Frontal, Central, Temporal, Parietal, Occipital and Insula). Automated sulci recognition was performed on Morphologist sulci models. Global and regional cortical surfaces and volumes were extracted, and values were harmonized for site and sex using pyComBat. Statistical analysis included t-test (mean volumes comparison), and normative scaling analysis based on a power law modeling between either cortical surfaces or cortical volumes and the Total Brain Volume (TBV, corresponding to the Cerebrum, the Cerebellum and the brainstem) in controls, providing 10th and 90th percentile curves. We ensured homocedasticity of residuals. We tested whether there was a signicant mean deviation of the patient group to the typical scaling using a shuffle-and-split procedure [6] and corrected for False Discovery Rate (FDR, Benjamini-Yekutieli). Effect size was measured as the mean of patients’ z-scores. An atypical sulcal pattern in the parieto-occipital region was observed in all 10 patients. Especially, major anomalies of the parieto-occipital sulcus led to its misidentification by Morphologist and to an incorrect segmentation of parietal and occipital lobes in 9 out of 10 patients. Central sulcus atypia was also observed in 8 out of 10 patients, but without misidentification (fig.1). As a result, we chose to group the parietal and the occipital lobes as one region for analysis, while segmentations of other regions appeared correct in comparison to controls. There was no difference in TBV between patients and controls (fig.2). Normative scaling analyses revealed a significant excess of global cortical surface in relation to TBV in patients (p-value < 10e-7, mean-z: 1.1) (fig.3). At a regional scale, a significant excess of surface in relation to TBV was observed in the temporal lobe (p-value < 10e-7, mean-z: 2.5) ) (fig.3), associated with a significant excess of volume (p-value < 10e-7, mean-z: 2.6) (fig.4). In addition to this result, qualitative observations showed major atypia in the shape of primary sulci on the outer surface of the temporal lobe, especially of the superior temporal sulcus, in all 10 patients (fig.1). An excess of volume in the parieto-occipital region was also found but with a lesser size effect (p-value=0.03, mean-z: 0.9) (fig.4). Theses preliminary results revealed an overall excess of cortical surface in BBSOAS patients, not explained by a normal TBV. This global increase seemed to be mainly related to an excess of cortical surface in the temporal lobe, associated to an oversizing (over-scaling) of its volume. This disproportionate scaling came with important modification in the folding pattern of large primary folds, in the temporal region but also the central and internal parieto-occipital ones, limiting our ability to separate between occipital and parietal lobes. Our results also highlight the potential of normative scaling analysis to study cortical development even in rare diseases. Work is still in progress to further characterize theses hemispheric anomalies in terms of surface and volume regional distribution (scaling), sulcal pattern, and to study their link with corpus callosum posterior thinning also described in BBSOAS patients [2].
Emma SOUCANE (Paris), Jérémy SADOINE, Ombline DELASSUS, Lucie HERTZ-PANNIER, Liubinka MIRAKOVSKA, Raphaelle MOTTOLESE, Laurence FAIVRE-DOURE, Michèle STUDER, Jean-François MANGIN, David GERMANAUD
13:30 - 15:00
#47574 - PG517 Effects of a Long-Term Exercise Intervention on Skeletal Muscle Metabolism in Aging Adults Assessed by Proton and Phosphorus Magnetic Resonance Spectroscopy Radka Klepochová1,2,3,4, Ivica, Just 3,4, Lucia Slobodová1,5, Petra Dubajová1,5, Miriam Dojčárová1.
PG517 Effects of a Long-Term Exercise Intervention on Skeletal Muscle Metabolism in Aging Adults Assessed by Proton and Phosphorus Magnetic Resonance Spectroscopy Radka Klepochová1,2,3,4, Ivica, Just 3,4, Lucia Slobodová1,5, Petra Dubajová1,5, Miriam Dojčárová1.
Healthy aging is increasingly recognized as a public health priority, with skeletal muscle health playing a central role in maintaining mobility, metabolic function, and overall quality of life in older adults (Tieland et al., 2017). Aging is associated with declining muscle mass, mitochondrial function, and metabolic flexibility, contributing to frailty and metabolic diseases. Regular physical activity, especially combining aerobic and resistance training, has been shown to counteract these changes by improving muscle functional capacity(Slobodová et al., 2022), mitochondrial efficiency and muscle bioenergetics (Hughes et al., 2018). Non-invasive magnetic resonance spectroscopy (MRS) enables in vivo quantification of skeletal muscle metabolites and mitochondrial function (Krššák et al., 2020; Meyerspeer et al., 2021). This study aimed to evaluate the metabolic effects of a long-term aerobic-strength exercise intervention in aging adults using both proton (1H) and phosphorus (31P) MRS.
Sixteen older adults (age 67 ± 8 years, BMI 28 ± 5 kg/m²) were randomly assigned into an aerobic-strength exercise group (n=10) and an active (stretching) control group (n=6). All participants underwent magnetic resonance (MR) measurements before and after the 9-month intervention. The exercise group participated in a supervised aerobic and strength training program three times 1 hour per week, complemented by dietary counselling (group educational sessions on monthly basis), while the active control group performed only light stretching exercises during the same 9-month period.
All MR measurements were performed in the morning following overnight fasting. Individuals were positioned supine with the right calf placed on a RF coil mounted on an ergometer dedicated for plantar flexion exercise inside of the 7T whole body MR system. Static 1H MRS of the gastrocnemius muscle was used to quantify acetylcarnitine, carnosine, and intramyocellular lipids (IMCL). Static 31P MRS assessed levels of phosphocreatine (PCr), phosphodiesters (PDE) (glycerophosphocholine (GPC) and glycerophosphoethanolamine (GPE)), inorganic phosphate (Pi), and phosphomonoesters (PME). A dynamic 31P-MRS protocol was used during and after a 6-minute submaximal plantar flexion exercise to evaluate phosphocreatine depletion (VPCr), phosphocreatine recovery (τPCr), maximum oxidative capacity (Qmax), and pH. There were no significant differences between the groups in any of the investigated metabolites at baseline. Following the intervention, the exercise group exhibited a trend toward increased acetylcarnitine (p=0.06), significantly lower GPC (p=0.02) and PDE (p=0.01), and elevated PME levels (p=0.04). Improvements in mitochondrial function were indicated from faster τPCr (p=0.05) and higher Qmax (p=0.05). Exercise performance, including delta PCr and end-exercise pH, was similar between the groups at both time points. No significant changes were observed in the control group. Our pilot results demonstrate that in aging individuals, exercise induces favorable changes in skeletal muscle metabolism detectable by advanced 1H and 31P MRS at 7T and dynamic exercise protocol in a rather small study population.
Trend to increased acetylcarnitine levels although not statistically significant (p = 0.06), may suggest improved mitochondrial substrate availability and possibly enhanced β-oxidation capacity(Bruls et al., 2019). The observed reductions in GPC and PDE are indicative of improved membrane turnover or reduced degradation, potentially reflecting healthier muscle cell membrane status, consistent with findings in trained older adults(Cikes et al., 2024). An increase in PME (p = 0.04) may indicate not only enhanced anabolic remodeling or regeneration of membrane structures but also altered metabolic activity. PME includes phosphorylated sugars involved in glucose transport and phosphorylation, as well as intermediates of glycolysis, suggesting broader changes in cellular metabolism. This supports the hypothesis that exercise promotes not only the preservation but also the renewal of muscle cellular architecture in aging tissue(Ziaaldini et al., 2017).
The faster phosphocreatine recovery time (τPCr) and increased maximal oxidative capacity (Qmax) reflect enhanced mitochondrial efficiency and ATP resynthesis, which are critical for muscle endurance and recovery. These improvements suggest that mitochondrial function is plastic even in later life, responding positively to regular training stimuli(Kemp et al., 1993). Exercise intervention in aging adults results in measurable improvements in skeletal muscle metabolism and mitochondrial capacity, as assessed by advanced 1H and 31P MRS at 7T. These findings underscore the importance of physical activity in promoting metabolic health and functional capacity in older populations.
Radka KLEPOCHOVÁ (Bratislava, Slovakia), Ivica JUST, Lucia SLOBODOVÁ, Petra DUBAJOVÁ, Miriam DOJČÁROVÁ, Pavol SZOMOLÁNYI, Viera LITVÁKOVÁ, Raynald BERGERON, Jozef UKROPEC, Barbara UKROPCOVÁ, Martin KRŠŠÁK
13:30 - 15:00
#47712 - PG518 REGIONAL T1 and T2 TRAJECTORIES in PEDIATRIC BRAIN DEVELOPMENT: DIFFERENTIAL MATURATION of LIMBIC and EXECUTIVE REGIONS.
PG518 REGIONAL T1 and T2 TRAJECTORIES in PEDIATRIC BRAIN DEVELOPMENT: DIFFERENTIAL MATURATION of LIMBIC and EXECUTIVE REGIONS.
Brain development from the fetal stage to adulthood involves dramatic and region-specific
changes. These changes are most rapid in the first few years of life[8]. The limbic system has a
vital role in emotional regulation and social communication. On the other hand, executive
functions are cognitive processes essential for controlling behavior in everyday tasks[4-10-19].
The study aims to investigate regional developmental trajectories by T1/T2 mapping across
childhood and adolescence.
The study analyzed the publicly available “Developmental Relaxometry Dataset” (80 healty
participants, 3-17 yr) hosted on OpenNeuro website. 36 subjects (17 toddlers, age 3–5 years;
19 adolescents, age 12–17 years) were randomly selected to form two age groups. Subjects
underwent whole-brain MP2RAGE scanning at 3T (MAGNETOM Prisma Fit, Siemens
Healthcare, Germany). T₁ maps were generated in MATLAB, fitting each voxel’s inversion
recovery signal (TI = 700, 2500 ms; TR = 5000 ms) via nonlinear least squares. We also
analyzed the T2 maps, provided in the dataset. Both maps were coregistered to the MNI
template in SPM12. The AAL atlas was used to define 11 regions of interest (ROIs): amygdala,
hippocampus, insula, parahippocampus, anterior cingulate cortex (ACC), ventromedial
prefrontal cortex (vmPFC),orbitofrontal cortex (OFC), fusiform, dorsolateral prefrontal cortex
(DLPFC), and composite limbic/executive regions. Mean T₁ and T2 values were extracted
within each ROI for all scans. Age trajectories were modeled by ordinary least-squares linear
regression of T₁ versus age. Unpaired two-sample t-tests compared children versus adolescents
per ROI, and paired t-tests compared limbic versus executive within each group. False-
discovery rate correction controlled for multiple ROI comparisons. Linear regressions revealed highly significant age-related decreases in T1 for the amygdala
(p<0.001), hippocampus (p<0.001), parahippocampus (p<0.001), ACC (p<0.001), composite
limbic (p<0.001), fusiform (p<0.001). The OFC showed significant increase (p<0.001). Slopes
in the insula, vmPFC, DLPFC, and the composite executive regions were non‐significant (all
p>0.05).
Between-group t-tests confirmed lower adolescent T₁ in the limbic composite (p<0.01),
amygdala (p<0.001), hippocampus (p=0.001), parahippocampus (p<0.001), ACC (p<0.001),
fusiform (p<0.001), higher OFC p<0.05), all survived FDR correction. Within-group paired t-
tests showed limbic network T₁ was significantly lower than executive T₁ in both children
(p<0.001) and adolescents (p<0.001).
In T2 regressions, significant age-related decreases observed in amygdala (p<0.001),
parahippocampus (p<0.001), and fusiform (p<0.001), but increase in th hippocampus (p<0.05).
All composite regions showed no change. Between t-test results were consistent with the
regression results, and no within group differences showed significance. This study mapped relaxation in limbic and executive networks from early childhood through
adolescence, revealing a heterogeneous maturation profile. Our findings align with previous
literature reporting rapid T₁ and T₂ relaxation changes in the first year of life, followed by
slower,region-specific maturation after age three[23-24-28]. In our study, all limbic regions except
the insula and fusiform demonstrated significant T1 decreases, whereas the amygdala,
parahippocampus, and fusiform showed T2 declines. Such early limbic lead is consistent with
the timeline of social-emotional development: children first develop parental bonding,
recognize and interpret others’ intentions[25], and acquire facial decoding via fusiform[20].This
early interaction lays the foundation for developing adaptive social behavior.
In contrast, T1 values in the OFC increased in adolescence, which seems like protraction, but
OFC and prefrontal cortex develop slowly[26]. We observed significant T2 increase in
hippocampus, likely arising from methodological differences in ROI detection: we applied
fully automated atlas while previous studies performed manual delineation. Myelination is a
dynamic process, it fluctuates asynchronously across regions[24].This asynchronous maturation,
limbic circuits maturing earlier than executive networks[22] may explain typical adolescent
behaviors: risk comprehension is intact, their elevated emotional responsiveness can dominate
decision‐making [18-19-21-22].
These observations align with normative patterns of myelination. Although absolute relaxation
times can vary with mapping protocols and MRI field strength, our analyses provide a robust
normative reference[22, 23]. Our relaxometry data establish clear, region specific measure: limbic circuits mature rapidly in
early childhood, while prefrontal networks continue myelinating through adolescence. These
quantitative T₁/T₂ metrics can be integrated into normative developmental models to enable
precise detection of atypical maturation trajectories in pediatric population.
Selin OZCAN (Istanbul, Turkey), Pinar S. OZBAY
13:30 - 15:00
#45431 - PG519 Functional connectivity breakdown between the Default mode and attentional networks as a ubiquitous mechanism of cognitive vulnerability during neurodevelopment and aging.
PG519 Functional connectivity breakdown between the Default mode and attentional networks as a ubiquitous mechanism of cognitive vulnerability during neurodevelopment and aging.
New computational approaches suggest that functional imbalance between key fMRI resting networks is critical for normal cognition and behavior, which would drive cognitive decline in neurodegenerative diseases. Specifically, the connectivity between the Default mode network (DMN) and the Attentional Networks such as the Dorsal (DAN) and Ventral attention networks (VAN), was identified as crucial for mental health.
We quantified the fMRI anti-correlation between DMN and DAN as a potential proxy for risk of neuropsychiatric disease in two studies A) In 2707 adolescents from the ABCB cohort https://nda.nih.gov/study.html?id=2248 , cognitive tests measuring intelligence were quantified along with demographic variables assessing risk for mental and neurologic diseases such as education level B) In another study fMRI and PET data from 183 elderly subjects with preclinical Alzheimer’s disease was analysed. We quantified the fMRI anti-correlation between the DMN and attentional networks using group ICA and dural regression in high-quality data from ADNI3. The between-network connectivity of the network of interest was quantified as the correlation between the time course of the ICA components of interest coming from dual regression analysis. The anti-correlation measures between the DMN and DAN were analysed along with PET tau, and PET Amyloid. Cognitive evaluation measures assessing general cognitive capabilities were the outcome variable I showed that an attenuated fMRI DMN-DAN anti-correlation is tightly correlated with general cognitive decline independently of p-tau pathology in elderly subjects and with general cognitive capabilities in adolescents. Together, these studies propose that DMN-DAN connectivity attenuation may represent a ubiquitous mechanism of cognitive vulnerability across the lifespan, from adolescence to aging and neurodegeneration One possibility is that the attenuation of the fMRI DMN-DAN anticorrelation would lead to brain network imbalance promoting spatial and temporal spreading of molecular changes. In this line, future studies should focus on understanding using multimodal neuroimaging and environmental factors such as childhood trauma, socioeconomic stress, and social isolation whether they could drive maladaptive connectivity between the DMN and attentional networks. This approach aims to understand the mechanism for neuropsychiatric diseases broadly, rather than only focusing on single genetic-molecular features as a source of cognitive decline and mental disease which hold as the mainstream proposed mechanism for neuropsychiatric diseases. As a possible future direction, one approach would be to attempt to build causality by focusing on longitudinal synergic interactions on large-scale studies between risk environmental and neuroimaging factors such as the fMRI DMN-attentional network imbalance as a possible feature Together these two studies shown that the DMN-DAN anticorrelation may represent an ubiquitous mechanism of cognitive vulnerability across the lifespan, from adolescence to aging and neurodegeneration
Diego Martin LOMBARDO VERA (Lac du Bourget)
13:30 - 15:00
#47660 - PG520 Characterizing normative trajectories of choroid plexus volume across the adult lifespan in healthy and diseased conditions.
PG520 Characterizing normative trajectories of choroid plexus volume across the adult lifespan in healthy and diseased conditions.
The Choroid Plexus (ChP) is a vascular structure within the brain's ventricular system that supports several physiological processes [1]. While prior studies have primarily investigated ChP volume (ChPV) in clinical populations—postulating a relationship between ChPV enlargement and neuroinflammatory mechanisms [2,3]—the contribution of normative biological variables (e.g., age, sex) and brain anthropometric measures (i.e. Total Intracranial Volume (TIV), Lateral Ventricle Volume (LVV)) to ChPV variability remains insufficiently characterized, partly due to limited healthy control (HC) sample sizes [4–6]. To address this gap, we introduce a normative modeling (NM) framework to delineate age-related trajectories of ChPV across the adult lifespan, aiming to advance its utility as a quantitative neuroimaging biomarker [7,8]. The NM was tested on two pathological cohorts of Multiple Sclerosis (MS) and Depression (DEP) subjects.
Data details are available in Table 1. The NM training dataset consisted of 3D T1-weighted MRI images from 1,036 HC sourced from the Cam-CAN [9] and HCP-A [10] datasets, restricted to a common age range (36–88 years). MRI quality was assessed using MRIQC v24.1.0 [11], and the ChP was automatically segmented using the fine-tuned ASCHOPLEX approach [12]. The validation dataset included 18 MS [12] and 26 DEP [13] subjects within the same age range, with ChPV also extracted with ASCHOPLEX. Lesion filling was applied to MS scans [14,15]. LVV and TIV were computed using FreeSurfer v7.1.1 [16]. Before building the Bayesian hierarchical NM, a preliminary statistical analysis on age, sex, dataset, ChPV, TIV, LVV, ChPV/LVV, and ChPV/TIV was performed on HCs using Python v3.12 and JASP [17] to identify which independent regressors should be included in the NM. This included independent samples t-tests (α = 0.05), Pearson’s correlations, and forward stepwise linear regressions predicting ChPV from age using different covariates. Model evaluation considered multicollinearity, statistical significance (p), explained variance (R², R² changes), and the Akaike Information Criterion (AIC). The NM was then trained and validated using Stan for R (v2.32.2) via the brms package (v2.22.0). ChPV was modeled with age, sex, TIV, and LVV as fixed effects, and dataset as a random effect: ChPV~Age+Sex+LVV+TIV+(1|Dataset). NMs used a Student’s t-distribution and were compared using Bayesian pseudo-R² and Bayes Factor (BF). The main output, Z-scores, quantified individual deviations from the normative posterior distribution. For HCs, Z-scores were computed using 5-fold cross-validation. For the MS and DEP cohorts, NM discriminative performance was assessed on Z-scores using ANOVA and the Area Under the Curve (AUC). The statistical analysis revealed no significant age differences between sexes. ChPV, LVV, and TIV differed significantly by sex (p<0.001), while Dataset-related differences were most pronounced for ChPV (Fig.1, Tab.2). Pearson’s correlation analysis showed statistically significant associations (p<0.001) between ChPV and age (0.35), LVV and age (0.57), ChPV/LVV and age (-0.55), ChPV and LVV (0.55), and ChPV and TIV (0.36). The optimal regression model included all variables as predictors, selected for the higher R² value (0.39) and lowest AIC. No multicollinearity was detected. Age, LVV, and sex emerged as the most influential predictors (R2 changes: 0.13, 0.13, 0.08). The NM including all covariates significantly outperformed simpler versions (BF>100, pseudo-R²=0.408) (Fig.2). Based on Z-scores distributions, ANOVA reported significant differences between groups (p<0.001), and the AUC was significantly greater than 0.5 (p<0.001) between HCs and both MS (0.69) and DEP (0.78) (Tab.2). This study presents a NM of ChPV across the adult lifespan, developed using a large cohort of HC and validated on two clinical populations. Based on high-quality segmentations from ASCHOPLEX, the model uses a hierarchical Bayesian framework to account for inter-scanner variability via partial pooling and shrinkage, enabling generalization to new scanners without the need for retraining. Statistical analysis confirmed the significant influence of age, sex, LVV, and TIV on ChPV, in line with prior findings [5]. The substantial effect of MRI acquisition protocols supports modeling the dataset as a random effect [18]. Importantly, the results show that normalizing ChPV by LVV or TIV does not improve interpretability. The selected NM explains approximately 40% of the variance in ChPV among HCs and effectively identifies individual deviations in clinical populations, consistent with existing literature [3,8,19]. The NM of ChPV across adulthood introduced in this study effectively identifies individual deviations in clinical populations with known ChPV alterations. The results underscore the model’s potential for clinical application and support further investigation of ChPV as a biomarker in both research and diagnostic contexts.
Valentina VISANI (Padova - Basel, Italy), Marco PINAMONTI, Alessio GIACOMEL, Manuela MORETTO, Maria Giulia ANGLANI, Agnese TAMANTI, Julia SCHUBERT, Federico TURKHEIMER, Alessandra BERTOLDO, Massimiliano CALABRESE, Mattia VERONESE, Marco CASTELLARO
13:30 - 15:00
#47838 - PG521 MRI Beyond Structural Narrowing in Spinal Stenosis: Quantifying Disc, Vertebral, and Age-Related Tissue Variation.
PG521 MRI Beyond Structural Narrowing in Spinal Stenosis: Quantifying Disc, Vertebral, and Age-Related Tissue Variation.
Lumbar spinal stenosis (LSS) is commonly diagnosed using MRI with qualitative or basic quantitative methods, such as dural sac cross-sectional area. While these clinical measures primarily reflect structural narrowing, they offer limited insight into tissue changes in discs and vertebrae, and their interpretation varies across institutions in the absence of standardized imaging criteria [1, 2].
Quantitative MRI-based metrics of disc and vertebral composition may capture additional tissue characteristics at clinically relevant levels, offering complementary information to current diagnostic practices. This study evaluates whether such quantitative MRI-based features can detect tissue differences in discs and vertebrae at spinal levels identified as most stenotic (index levels) or selected for surgical decompression in LSS.
The cohort included 350 patients with LSS (mean age 66.7 years; 184 male) from the multicenter Norwegian Degenerative Spondylolisthesis and Spinal Stenosis (NORDSTEN) study. Preoperative sagittal T2-weighted MR images were analyzed for discs and vertebrae between L1 and L5, using the full image volume. For each spinal level, the analysis covered the entire intervertebral disc and the full superior and inferior vertebral bodies. Tissues were segmented using MONAI, and disc spatial variation was assessed by dividing each disc into five equal-width anterior–posterior subregions with MATLAB.
Quantitative features included mean signal intensity (SI), standard deviation (SD), disc degeneration (Δµ [3, 4]), and vertebral entropy (signal disorganization). Images were normalized to the cerebrospinal fluid SI to reduce inter-scan variability.
Statistical differences were tested between index and non-index levels, and between surgical and non-surgical levels. Associations with age were evaluated using Pearson’s correlation coefficient (r). SI, SD, and Δµ tended to be lower in discs at index and surgical levels, with statistically significant differences particularly in central subregions (2–4) of the nucleus pulposus (p < 0.04; Table 1).
SD and entropy were generally higher for vertebrae adjacent to surgical levels than at non-surgical levels, particularly in the lower spine (p < 0.02; Table 1). In contrast, vertebrae adjacent to index levels tended to show lower entropy in caudal segments. Entropy also showed a stronger association with index level than SI, SD, or Δµ, with significant correlations at multiple levels (|r| ≥ 0.13, p < 0.01; Table 1, Figure 1).
At most spinal levels, disc degeneration increased with age, reflected by decreasing Δµ values, except at L4/L5, where no significant correlation was found. Vertebral entropy consistently decreased with age across all levels (r ≤ −0.21, p < 0.01), with no level-specific dependency (Figure 2). LSS patients showed measurable tissue changes at clinically relevant spinal levels. Metrics in the nucleus pulposus showed the strongest differences at index and surgical levels, suggesting a central role in LSS. While these patterns are consistent with known disc degeneration mechanisms, our results quantitatively confirm this association with LSS. Nucleus degeneration may reduce disc height and alter load distribution, increasing stress on surrounding structures and promoting joint degeneration and ligament thickening, both contributors to stenosis. Vertebral metrics also differed, with lower entropy at index levels, especially in the lower spine, while entropy and SD were higher at surgical levels. These patterns suggest that marrow organization varies along the spine and may reflect disorganization or compositional changes relevant to LSS.
The lack of age-dependent disc degeneration (Δµ) at L4/L5 suggests that mechanical or anatomical factors may contribute more to degeneration at this segment. This supports the hypothesis that age alone does not explain lumbar tissue changes. Vertebral marrow heterogeneity may therefore offer additional insight. Entropy captures this variation, which has also been described using other imaging methods [7, 8], though its application remains largely unexplored. In this study, entropy consistently decreased with age across vertebral levels, possibly reflecting more homogeneous marrow due to fatty infiltration or evolving Modic transformations. While Modic changes are often visible, entropy may provide an objective measure of subtle marrow variations not captured by conventional imaging. Quantitative MRI metrics, especially Δµ and vertebral entropy, identified tissue changes in the nucleus pulposus and adjacent vertebral marrow at levels affected by LSS. Entropy captured marrow characteristics beyond age-related degeneration, and findings suggest that current assessment practices might overlook clinically important variations. These imaging features warrant further investigation and may offer synergistic value in future AI-driven analyses to inform and improve treatment strategies for LSS patients.
Christian WALDENBERG (Gothenburg, Sweden), Ella NILSFORS, Erland HERMANSEN, Hanna HEBELKA, Hasan BANITALEBI, Helena BRISBY, Kari INDREKVAM, Kerstin LAGERSTRAND
13:30 - 15:00
#47002 - PG522 Regional cortical folding alterations in craniostenosis : an MRI-Based sulcation analysis.
PG522 Regional cortical folding alterations in craniostenosis : an MRI-Based sulcation analysis.
Craniosynostosis is a rare congenital disorder caused by the premature fusion of one or more cranial sutures, which can affect skull shape and brain development [1]. While its impact on overall brain morphology is known, effects on cortical folding remain poorly understood. This study aimed to quantify regional sulcation patterns in different craniosynostosis subtypes (Apert, Crouzon, Muenke, and non-syndromic) using MRI-based surface analysis.
The study included 65 patients with craniosynostosis, imaged at Hôpital Necker-Enfants Malades (mean age=5.46 years; 38 males, 25 females). The patient group comprised 33 individuals with Crouzon syndrome (16 pre-operative and 7 post-first surgery), 4 pre-operative Apert patients, 6 post-operative Muenke patients, and 23 post-operative non-syndromic patients. A control group of 130 typically developing children (mean age=5.99 years; 72 males, 58 females) was also included. Control data were obtained from Hôpital Necker-Enfants Malades, the Baby Connectome Project [2], and Neurospin, with 24.6% of controls scanned at Necker to ensure partial site matching. All participants were imaged with high-resolution 3D isotropic T1-weighted MRI scans. Images were processed using the Morphologist pipeline from BrainVISA [3] to extract cortical sulcal surfaces from eight hemispheric regions, defined according to cranial sutures (anterior and posterior coronal, temporal, and posterior lambdoid, on both mesial and lateral surfaces shown in Figure 1). The hemispheric hull surface was also computed. To reduce inter-site and sex-related variability, the cortical surfaces were harmonized using ComBat. A sulcation index was calculated for each region as the ratio between the regional sulcal surface area and the corresponding hemispheric hull surface area. A sulcation index was also calculated for each hemisphere. Normative developmental trajectories were modeled in the control group using power-law growth functions. Patient data were compared to this normative model to identify atypical sulcation, defined as values falling below the 10th or above the 90th percentile (Fisher's test). Statistical comparisons of residuals were performed using shuffle and split tests to assess group-level differences. No significant differences in hemispheric sulcation were observed between any disease group and controls. Regarding regional sulcation, age-adjusted comparisons with controls revealed the following findings. In Crouzon syndrome, increased sulcation was observed in the temporal region (corrected p-value=10^-5 for the left hemisphere, 0.01 for the right) and in the posterior lambdoid region (left hemisphere, corrected p-value=10^-5 for the right hemisphere) but a decreased sulcation in the left hemisphere (corrected p-value=10^-2) . In Apert syndrome, no significant differences were found, likely due to the small sample size. In Muenke syndrome, decreased sulcation was observed in the anterior coronal region (corrected p-value=0.04 for the left hemisphere, 0.007 for the right), posterior coronal regions (corrected p-value=0.03 for the left hemisphere, 0.007 for the right), and an increased sulcation for the temporal (left hemisphere, corrected p-value=10^-8) and internal temporal regions (left hemisphere, corrected p-value=10^-8). Finally, in the non-syndromic group, increased sulcation was observed in the posterior coronal region (left hemisphere, corrected p-value=0.01). Our findings reveal no significant differences in hemispheric sulcation between any disease group and controls. However, significant regional differences in sulcation patterns across craniosynostosis syndromes, with Crouzon and Muenke patients showed alterations compared to controls. Increased sulcation in regions such as the temporal and posterior lambdoid in Crouzon syndrome, as well as the anterior and posterior coronal in Muenke syndrome, suggests that these patterns may reflect neurodevelopmental differences associated with cranial malformations and their respective management. In Apert syndrome, no significant differences were observed, likely due to the small sample size, highlighting the need for larger cohorts to better characterize sulcation patterns in this group. For Crouzon syndrome, the variability observed in sulcation patterns may be related to the genetic and suture closure diversity of the mutations involved. The non-syndromic group exhibited increased sulcation in the posterior coronal region, although the underlying cause of this effect remains unclear. Regional sulcation alterations were identified in Crouzon and Muenke syndromes, despite the absence of global gyration differences. Regional sulcation alterations were identified in Crouzon and Muenke syndromes, despite preserved global gyration. These results confirm that localized cortical folding changes can occur in craniosynostosis and may reflect underlying neurodevelopmental variability specific to each syndrome, beyond the effects of skull shape alone.
Ombline DELASSUS (Paris), Lucas CHOLLET, Barbara YOUNGUI, Giovanna PATERNOSTER, Roman Hossein KHONSARI, David GERMANAUD, Jean-François MANGIN
13:30 - 15:00
#47565 - PG523 Case Study on Quantitative MRI in a 71-Day-Old Rat Brain with Cortical Malformation.
PG523 Case Study on Quantitative MRI in a 71-Day-Old Rat Brain with Cortical Malformation.
Quantitative MRI (qMRI) is a non-invasive technique that investigates the alterations in the microstructure of the tissues by estimating relaxation times such as T1, T2, and T2*, and derived metrics like R2* [1]. Quantitative Susceptibility Mapping (QSM), is another qMRI technique that estimates the magnetic susceptibility of the tissues and gives information about iron accumulation, calcium, and myelin content [4]. These parameters are sensitive to changes in water content, iron accumulation, and myelination. Quantitative MRI can be used to detect biomarkers that characterize neuropathology [2]. Rodent models are used in preclinical studies due to their genetic similarities to humans. Rodent models can be used for aging studies due to their short lifespan, which allows for efficient data collection and longitudinal studies [3]. In this study, we aimed to use qMRI techniques to analyze a 71-day-old rat with a cortical malformation and compare its quantitative measurements, including those from adjacent and periventricular parenchyma, to those of age-matched healthy controls.
The data is acquired from an ongoing preclinical aging study. The study includes eighteen female Wistar rats (mean age: 74.2 ± 8.2 days; weight: 178.2 ± 11.6 g) scanned using a 7 Tesla preclinical MRI scanner (MR Solutions Ltd., UK). T1-weighted (T1w, Fast Spin Echo (FSE), TR/TE: 1000/11 ms, FA: 90°, Resolution: 0.125 × 0.125 × 0.8 mm³), T2-weighted (T2w, FSE, TR/TE: 2500/40 ms, FA: 90°, Resolution: 0.125 × 0.135 × 0.8 mm³), Multi-Echo Multi-Slice (MEMS, TR/TE: 4000/150 ms, FA: 90°, Resolution: 0.16 × 0.31 × 1 mm³), and Multi-Gradient Echo (MGE, TR: 1620 ms, FA: 60°, Resolution: 0.36 × 0.36 × 0.36 mm³, Min/Max TE: 4/21.12 ms, 9 echo times). The FLAIR sequence was also acquired for the case with cortical malformation. These sequences are used to calculate the quantitative parameters, including T1, T2, T2*, and R2* maps.
One Wistar female rat (71 days old) spontaneously exhibited a schizencephaly-like cleft extending from the cortical surface to the ventricles, establishing communication with the ventricular system in the right hemisphere. The cleft demonstrated CSF-equivalent signal intensity across all imaging sequences (Figures 1,2). It is located across both cortical and subcortical regions, including parts of the somatosensory cortex, insular cortex, and deep white matter. Associated findings include right-sided asymmetric hydrocephalus and inward displacement of the corpus callosum. This animal was selected for case analysis, and the region with the cleft was analyzed. Five age-matched female rats with no visible abnormalities were selected as controls for comparison.
For the quantitative analysis, first, skull stripping was done on the subjects using an in-house UNet model (presented on ISMRM 2025). Then, voxelwise maps of relaxation parameters were computed using custom codes in Python to fit standard signal models. Voxel-wise relaxation maps (T1, T2, T2*, and R2*) were computed by fitting standard signal models for each sequence: inversion recovery for T1 (IR-FLASH), multi-echo spin echo for T2 (MEMS), and multi-echo gradient echo for T2* and R2* (Figure 3). SEPIA (v1.2.2.3) [6] in MATLAB was used to generate QSM. GRE phase and magnitude data were processed using Laplacian unwrapping (STI suite) [9], VSHARP [8] for background removal, and star-QSM [7] for dipole inversion.
After generating the maps, they were registered to the Sigma rat brain atlas [5]. With guidance from a radiologist, the corresponding anatomical labels were then used to identify regions containing the lesion. The quantitative values extracted from the cleft region showed marked deviation from age-matched healthy controls. The T2 values in the cleft region were notably higher (171.33 ms, case; 59.22 ± 1.04 ms, controls). Similarly, T1 relaxation time was prolonged (2582.36 ms, case; 1566.08 ± 33.12 ms, controls). The T2* value was also increased (4.48 ms, case; 0.07 ± 0.0035 ms, controls), while R2* showed a reduction (10.21, case; 14.90 ± 0.51, controls). Lastly, QSM values were slightly lower in the cleft (0.0019 ppm, case; 0.0023 ± 0.0007 ppm, controls). In contrast, values from unaffected brain regions were consistent with age-matched healthy controls. Quantitative maps revealed increased T1, T2, and T2* values in the cleft, indicating increased water content and reduced tissue complexity and myelination. On the other hand, R2* decreased, suggesting decreased magnetic susceptibility effects. QSM values were slightly decreased, supporting the presence of CSF-like fluid and altered microstructure. This case study demonstrates how qMRI enables the detection and characterization of brain abnormalities in preclinical models.
Acknowledgments: This research is supported by TÜBİTAK 1004 Grant (No. 22AG016).
Leen HAKKI (Istanbul, Turkey), Öykü YEŞILOĞLU, Belal TAVASHI, Uluç PAMUK, Oğuzhan HÜRAYDIN, Esin ÖZTÜRK IŞIK, Pınar Senay ÖZBAY
13:30 - 15:00
#47688 - PG524 Volumetric MRI Analysis in 3-Month-Old Wistar Rats: A Baseline for Longitudinal Studies.
PG524 Volumetric MRI Analysis in 3-Month-Old Wistar Rats: A Baseline for Longitudinal Studies.
Aging is a natural process, and with the increase in human lifespans, research into the effects of normal aging on brain structures and volume has become increasingly important [1]. Magnetic resonance imaging (MRI) is a commonly used noninvasive method in the literature for studying aging. Rodent models are widely used in preclinical studies because of their genetic similarities to humans and their short lifespan, allowing for efficient data collection and longitudinal studies [2]. This enables researchers to analyze the aging effect within the same individuals at different time points.
Volumetric analysis of brain structures is important in characterizing age-related morphological changes. Regions such as the striatum, thalamus, corpus callosum, subiculum, and hypothalamus are known to undergo structural alterations with age [3]. Furthermore, measures of gray matter (GM), white matter (WM), cerebrospinal fluid (CSF), and total intracranial volume (TIV) provide an overview of overall brain structure [4].
As part of an ongoing aging study, we performed a baseline volume analysis on a group of same-aged Wistar rats (~3 months old) using anatomical MRI images. While the full longitudinal data is not yet available, this first time point provides a reference for key brain regions and tissue types. These measurements will be used to track volume changes related to aging in future time points. In addition, tissue probability maps will be used for voxel-based morphometry (VBM) to examine whole-brain differences.
This study includes eighteen same-aged female Wistar rats (mean age: 74.2 ± 8.2 days; weight: 178.2 ± 11.6 g) scanned using a 7 Tesla preclinical MRI scanner (MR Solutions Ltd., UK). T1-weighted (T1w, Fast Spin Echo (FSE), TR/TE: 1000/11 ms, FA: 90°, Resolution: 0.125 × 0.125 × 0.8 mm³), T2-weighted (T2w, FSE, TR/TE: 2500/40 ms, FA: 90°, Resolution: 0.125 × 0.135 × 0.8 mm³), Multi-Echo Multi-Slice (MEMS, TR/TE: 4000/150 ms, FA: 90°, Resolution: 0.16 × 0.31 × 1 mm³), and Multi-Gradient Echo (MGE, TR: 1620 ms, FA: 60°, Resolution: 0.36 × 0.36 × 0.36 mm³, Min/Max TE: 4/21.12 ms, 9 echo times). T2w images were used for the volumetric analysis.
To prepare the data for analysis, the skull stripping was done using an in-house U-Net model with a 19-convolutional layer (VGG19) encoder developed especially for rat MRI scans (presented on ISMRM 2025) (Figure 1). For generating tissue probability maps (TPM), first, the volumes were affinely registered to the Sigma Brain Atlas [5] using ANTsPy [6], followed by segmentation with SPM8 [7].
For regional volume calculation, inverse nonlinear registration was performed, where the atlas was nonlinearly warped to each subject’s native space, and the volumes of striatum, corpus callosum, thalamus, hypothalamus, and subiculum were assessed. The TIV was 3557.7 ± 63.8 mm³. Among the tissue probability maps, GM showed the highest volume (1580 ± 31.2 mm³), followed by the WM (994.7 ± 32.5 mm³) and cerebrospinal fluid (CSF: 983.0 ± 18.2 mm³). The regional volume measurements showed that the largest structure was the striatum (97.6 ± 5.6 mm³), followed by the corpus callosum (76.6 ± 5.2 mm³), thalamus (57.8 ± 3.8 mm³), hypothalamus (33.2 ± 3.2 mm³), and subiculum (16.2 ± 1.2 mm³). The box plots of the distribution of regional volumes and TPM-based tissue volumes are shown in figures 3 and 4. In this study, we developed and implemented a preprocessing and analysis pipeline to support longitudinal analysis of age-related brain changes in Wistar rats. The current work focuses on the baseline time point, providing volumetric measurements of global brain tissues and selected regions of interest, which include GM, WM, CSF, and key brain regions known to be affected by aging. Our pipeline combines automated brain extraction using a deep learning model, atlas-based registration with ANTs, tissue segmentation with SPM8, and regional volume estimation via inverse warping of the atlas. This approach can be used for a consistent longitudinal assessment of regional volumes in the same animals. This study provides baseline volumetric data for early adult Wistar rats and establishes a pipeline for future longitudinal analysis. The results presented offer a reference for tissue maps and regional brain volumes, supporting future investigations for detecting structural changes related to the aging process.
Acknowledgments: This research is supported by TÜBİTAK 1004 Grant (No. 22AG016).
Leen HAKKI (Istanbul, Turkey), Belal TAVASHI, Uluç PAMUK, Oğuzhan HÜRAYDIN, Esin ÖZTÜRK IŞIK
13:30 - 15:00
#46433 - PG525 Quantitative MRI of exogenous Pulmonary Surfactant Distribution: Ex-vivo and In-vivo Studies.
PG525 Quantitative MRI of exogenous Pulmonary Surfactant Distribution: Ex-vivo and In-vivo Studies.
Respiratory Distress Syndrome (RDS) remains a major cause of morbidity and mortality in preterm infants, primarily due to pulmonary surfactant deficiency. Administration of exogenous surfactant is a cornerstone of neonatal intensive care. Furthermore, pulmonary surfactant shows promise as a drug delivery vehicle for targeting lung diseases such as inflammation and infections (1,2). This study evaluates the potential of Magnetic Resonance Imaging (MRI) for imaging and quantifying the distribution of exogenous surfactant in preclinical rabbit models, both ex-vivo and in-vivo, mimicking the pulmonary characteristics of preterm infants.
Ex-vivo experiments were conducted on isolated rabbit thoraxes (N=6) ventilated under pleural depression and Positive End Expiratory Pressure conditions (3). In-vivo studies used anesthetized 6-week-old rabbits (1 kg body weight, N=6). In both cases, 2 mL of Curosurf (Chiesi Farmaceutici) surfactant solution mixed with 0.2 mL of Dotarem (Guerbet) contrast agent solution were instilled in the lungs using the clinical standard Less Invasive Surfactant Administration approach (4) (Fig. 1a-b, Fig. 2a). 3D UTE (Ultra-short Echo time) MRI scans were acquired on a 3T whole-body magnet (Vantage Centurian, Canon Med. Sys.). T1 maps of the surfactant were generated using a Variable Flip Angle approach (5) and then concentration maps were generated. Central airways, peripheral lung regions and Total lung volume were segmented with the nnU-Net deep learning framework (6) for ex-vivo images and manually for in-vivo images. In ex-vivo experiments, MRI signal enhancement reached up to 1900% (Fig. 1c–d), with 84% of the instilled surfactant distributed to distal lung regions, occupying 32.5% of the total lung volume. In-vivo imaging showed signal enhancements up to 1700% (Fig. 2b–c), with the surfactant distribution remaining stable over a one-hour period and no signal enhancement observed outside the pulmonary regions. Gd³⁺ concentration maps (Fig. 3a, 3d) revealed surfactant presence along the walls of the large airways and demonstrated inhomogeneous distribution between the left and right lungs in both ex-vivo and in-vivo experiments.
Segmentation of the central airways and lung volumes (Fig. 2b–c, 2e–f) enabled quantitative assessment of the contrast-enhanced surfactant distribution, showing variation along the ventral to dorsal direction with lungs in the supine position during surfactant administration and MRI studies. (Fig. 3a, 3c), as well as notable asymmetry between the right and left lungs (Fig. 3b, 3d). These results demonstrate the feasibility of using MRI to visualize and quantify the distribution of the surfactant within the lungs both in-vivo and ex-vivo. The marked inhomogeneity observed between the left and right lungs suggests that surfactant delivery can be uneven, potentially influenced by factors such as anatomy or the animal's position during administration. The lack of signal enhancement in other tissues or blood vessels indicates that the contrast agent remained co-localized with the surfactant into the airspaces. Additionally, the minimal change in surfactant distribution over time suggests stability of the administered surfactant within the lung during the imaging period. This study validates the use of MRI for the quantitative evaluation of pulmonary surfactant distribution in both ex vivo and in vivo models. The proposed approach is currently being used to improve surfactant administration techniques for premature infants, and it also holds promise for evaluating the distribution of therapeutic agents using surfactant as a drug carrier. Overall, this new method offers broader implications for optimizing drug delivery and enhancing treatment strategies.
Oumaima MARFOUK (Bordeaux), Rémy GÉRARD, Ghalia KAOUANE, Lara LECLERC, Fanny MUNSCH, Bei ZHANG, Stéphane SANCHEZ, Noël PINAUD, Mickaël FAYON, Sophie PÉRINEL-RAGEY, Jérémie POURCHEZ, Eric DUMAS DE LA ROQUE, Yannick CRÉMILLIEUX
13:30 - 15:00
#47891 - PG526 AI-based volumetric analysis of brain oedema in experimental cerebral malaria.
PG526 AI-based volumetric analysis of brain oedema in experimental cerebral malaria.
Cerebral malaria (CM) is the most lethal complication of Plasmodium falciparum infection that occurs in 1-2 % of cases and affects mostly children under 5 years in sub-saharan Africa. This encephalopathy that has a 15-20% fatality rate is characterized by seizures, altered consciousness and coma. Brain swelling is considered as a major predictor of death [1]. The mouse model of CM obtained with P. berghei ANKA (PbA) is a clinically relevant model of the disease characterized by massive brain swelling [2]. The aim of this study was to identify the brain structures most affected by vasogenic oedema in both male and female mice which could lead to a better understanding of the neurological sequelae in survivors. To this end, we imaged the mice before CM induction and at the peak of the disease using anatomical MRI. Images were analysed using a tool developed in-house based on AI for the automatic segmentation of brain structures.
27 male and 15 female C57Bl/6J mice aged 8 to 10 weeks were imaged before inoculation with 2x10^6 erythrocytes parasitized with PbA and at the peak of CM (d5-6 post inoculation). The clinical monitoring included the assessment of neurological signs, body weight, temperature and parasitaemia. MRI experiments were carried out on a Bruker AVANCE 500 WB system @11.75 T equipped with a volume T/R coil for the mouse brain and a heating blanket, under isoflurane anaesthesia (1-2%/air) and physiological monitoring. Images were acquired using the RARE sequence: TR/TE: 5000/9.21 ms, RARE factor 8, 4 av, FOV 15x15 mm2, 194x194 matrix, 31 contiguous slices of 0.5 mm. The images were segmented using the nnU-Net framework [3] and a training database of 18 2D mouse brain MR images manually segmented by experts. The automatic segmentation produced 28 parcels including the brain mask. For each mouse, the volume fraction (FV) of each structure with respect to the whole brain volume was calculated as: FV = Vstr/ Vbrain.ctrl. Vstr is the volume of the structure at each time point and Vbrain.ctrl the volume of the whole brain before CM induction. For each mouse, the volume variation of each structure caused by the disease (delta was calculated as: delta = [(Vstr.peak - Vstr.ctrl)/ Vstr.ctrl] * 100. Statistics: two-way ANOVA followed by Šidák’s multiple comparison test, significance: p<0.05. 93% of the males and 74% of the females developed CM. Twelve males and two females reached a humane endpoint before imaging and had to be withdrawn from the study. The weight loss appeared from day 2 after PbA inoculation in males and a day later in females, while the neurological signs pathognomonic of the severe stage of CM occurred between d5 and 6. Hypothermia, a well-known feature of murine CM resulted in an average drop in body temperature of 13% at d5-6 in both sexes. Parasitaemia at d5 was between 5 and 16% with no significant difference between males and females. Both sexes presented significant brain swelling on MRI particularly visible along the dorsoventral axis and at the level of the cerebellum whose fissures were less distinguishable. Oedema was associated with micro-haemorrhages that were more numerous in males than in females. The total brain volume increased by 4-5% at the peak of CM (males: Vctrl = 474.2 ± 14.4 mm3, Vpeak = 494.7 ± 17.6 mm3; females: Vctrl = 467.7 ± 14.2 mm3, Vpeak = 491.2 ± 11.6 mm3). Both males and females presented significant augmentation of the volume of the pons, cortex, and striatum (Fig 1). Cortex was responsible for ca 50% of the increase (average cortical increase of 9.9 ± 5.6 mm3 in males and 11.0 ± 8.8 mm3 in females) (Fig 1-2). Other volume changes were specific to one sex. Males presented a significant increase of the cerebellum, ventricles, inferior colliculus and periaqueductal gray, and a reduction of the superior colliculus whereas females presented a significant increase of the midbrain. The volumetric study of 27 brain structures enabled the identification of structures that are equally affected in both sexes such as cortex, pons and striatum, as well as sex-dependent differences in other structures including the superior colliculi, cerebellum or midbrain. A downregulation of pro-inflammatory cytokines by oestradiol in females and the immunosuppressive effect of testosterone in males could partly explain the differences between males and females [4]. CM causes a cerebral syndrome that affects females and males differently. These sex-linked differences, reflected in some regional prevalence studies in endemic areas, also appear at the peak of experimental CM at the level of the clinical signs and the distribution of the oedema in specific brain structures. This is the first description of the regional development of oedema in CM and the first imaging-based demonstration of sex differences in CM. This study could help advance the understanding of brain damage in CM according to sex and encourage the search for sex-specific adjunct therapies.
Alicia COMINO GARCIA-MUNOZ (Marseille), Isabelle VARLET, Oumaïma MARFOUK, Constance MICHEL, Ludivine GUYOT, Emilien ROYER, Teodora-Adriana PERLES-BARBACARU, Angèle VIOLA
13:30 - 15:00
#47869 - PG527 Predicting histological features in human sclerotic and non-sclerotic hippocampi using frequency-dependent multidimensional diffusion-relaxation MRI.
PG527 Predicting histological features in human sclerotic and non-sclerotic hippocampi using frequency-dependent multidimensional diffusion-relaxation MRI.
Magnetic resonance imaging (MRI) is key for evaluation of patients diagnosed with drug-resistant epilepsy[1]; however, current clinical MRI methods lack specificity to distinguish histopathological changes[2] associated with the development of epileptogenic tissue, which significantly impacts the accuracy and effectiveness of surgical resection.
Frequency-dependent multidimensional diffusion-relaxation correlation MRI (ωMDR-MRI) offers a novel analysis framework to disentangle sub-voxel information, providing specificity on heterogeneous cellular environments[3].
In this work, we performed a regression analysis to predict cell morphological features of sclerotic and non-sclerotic hippocampi using the diffusion and relaxation properties provided by ωMDR-MRI at high resolution in five patients with focal drug-resistant epilepsy. This approach offers a new analytical method to improve the accuracy of non-invasive studies of the hippocampal state in these patients.
Five patients with focal drug-resistant epilepsy underwent anterior temporal lobectomy. Four hippocampi were diagnosed as hippocampal sclerosis (HS) and the remaining one was non-sclerotic. A portion of the resected hippocampi were imaged using a 11.7 T vertical Bruker scanner (gradient strength 3000 mT/m). The multidimensional images were obtained using a segmented Spin Echo-Echo Planar Imaging (SE-EPI) sequence customized to acquire diffusion with general gradient waveforms. The multidimensional acquisition consisted of 352 images with isometric voxel size of 150 μm3 varying b-value (0.436-9.21·109 sm–2), centroid frequency ωcent/2π (32-252 Hz), normalized anisotropy bΔ (–0.5, 0, 0.5, and 1), orientation (Θ,Φ), repetition time TR (800-3500 ms), and echo time TE (11-65 ms). We obtained the parameter distributions derived from ωMDR-MRI analysis (Diso, DΔ2, R1, R2, Δ????/2π Diso and Δ????/2π DΔ2) with their corresponding per-voxel means and bin-resolved maps as in previous studies (Fig. 1).
Cell bodies were segmented from two Nissl micrographs (Fig. 2A-C) per sample using a fine-tuned version of the vision transformer Segment Anything Model (SAM)[4]. Then, we extracted cell shape measurements from each detection, such as the area, minimum and maximum diameter, and solidity (Fig. 2D-F). The segmented slices were down-sampled to match the MRI voxel size creating histological reference quantitative maps (Fig. 2G-I). Each pair of segmented slices per subject were aligned and registered to its corresponding MRI slice (Fig. 3). Then, we performed a regression analysis using the random forest (RF) approach[5] and obtained the training R2. Separated RF models were trained with different MRI parameters (Fig. 1): Diffusion tensor imaging (DTI), 2 maps; ωMDR per-voxel means, 6 maps; and ωMDR bin-resolved, 18 maps. We evaluated the prediction accuracy and performance with leave-one-out cross-validation (LOO-CV) and metrics such as Q2, and MSE[6]. Fig. 4 shows a comparison of the three regression models on the histology target variables of cell density and area. We found in both representative sclerotic and non-sclerotic hippocampi that ωMDR bin-resolved predict more accurately the ground truth (shape measurements derived from cell segmentation). Regions with high cell density, such as the granule cell layer were underrepresented in the DTI predictions, with improvements seen in the ωMDR bin-resolved predictions. The opposite effect was present in regions with lower cell density, such as the subiculum, where the predictions overrepresent the cell count.
The overall performance of the RF regression model (Table 1) showed that DTI has the lowest area prediction accuracy among all variables, with an R² value of 0.57. An improvement of R² was obtained (R²=0.79) using ωMDR per-voxel means. The highest value of R² = 0.86 was reached with ωMDR bin-resolved predictions. A similar trend was observed for the remaining target variables. The LOO-CV metric Q² shows that the predictions remained positive for all target variables across all three models, except for cell density. ωMDR-MRI better predicts morphological cell features extracted from the Nissl staining compared to DTI. This demonstrates the relevance of frequency-dependent [7–9] and bin-resolved tensor-valued encoding diffusion[10–12] to better characterize the cell morphology. While the relaxation properties might have also weight in differentiating water populations within each voxel[13]. Our results showed that even with a limited dataset from five hippocampi, good regression performance was achieved; however, it is needed to explore the regression with additional MRI and histological hippocampi slices and more resected hippocampi. The use of multiple parameters obtained from ωMDR-MRI on ex vivo resected hippocampal tissue to predict histological results presents new opportunities to in vivo translation which could enhance diagnostic specificity, surgery planning and monitoring of patients with drug-resistant epilepsy.
Omar NARVAEZ (Kuopio, Finland), Jenni KYYRIÄINEN, Maxime YON, Sara GRÖHN, Saana ELAY, Tuomas RAURAMAA, Mastaneh TORKAMANI, Arto IMMONEN, Ville LEINONEN, Henri ERONEN, Reetta KÄLVIÄINEN, Daniel TOPGAARD, Tarja MALM, Jussi TOHKA, Olli GRÖHN, Alejandra SIERRA
13:30 - 15:00
#47322 - PG528 The effect of electroconvulsive therapy and transcranial magnetic stimulation on brain volumes and perfusion in depression.
PG528 The effect of electroconvulsive therapy and transcranial magnetic stimulation on brain volumes and perfusion in depression.
Major depressive disorder (MDD) is characterized by persistent low mood, anhedonia, and functional impairment without a history of mania or hypomania [1][2]. In bipolar disorder, depressive episodes are phenotypically similar but occur in individuals with a history of manic or hypomanic episodes. [3]. Electroconvulsive Therapy (ECT) and Transcranial Magnetic Stimulation (TMS) are treatment options for treatment-resistant depression [4][5] and bipolar disorder depression [6]. Volume changes in anatomical structures such as the hippocampus are consistently reported following ECT [7]. There are fewer studies of volume change after TMS, but volume changes in the hippocampus have been reported [8]. Studies on brain perfusion, or Cerebral Blood Flow (CBF), when assessed by Positron Emission Tomography (PET) and Single-Photon Emission Computed Tomography (SPECT) have shown changes in relative CBF both during and after ECT [9].
The aim of the current study was to investigate a potential association between changes in brain MR perfusion using arterial spin labelling and volume changes in selected brain structures. The hypothesis was that the ECT and TMS treatment will lead to increased volume in the selected brain structures and that the same structures will experience a change in perfusion.
The MRI images were acquired on a GE Discovery MR750w 3.0T, using 3D MP-RAGE (TE/TR/TI=3.1/7.4/1060ms, 1mm3) for the T1 images and 3D-ASL (pcASL, TE/TR=10.5/4888ms, 1.875x1.875x4 mm3) for the perfusion images. FreeSurfer [10] and FSL BASIL [11] were used to analyze the images. FreeSurfer was used for image segmentation and volume quantification, while FreeSurfer and FSL BASIL were used for perfusion quantification. At the time of analysis, the data consisted of 28 participants from the GEMRIC study [12], where 9 had received ECT, 10 had received TMS and 9 were healthy controls (Figure 3). MRIs were collected at five timepoints for the patients: 1 hour before, 1 hour after, 14 days after, 3-8 weeks after and 7-8 months after first treatment, and four timepoints for the healthy controls over the same time span, lacking the MRI at 3-8 weeks, as described in the study protocol [13] (Figure 4). The results showed a statistically significant volumetric increase from scan 1 (before treatment) to scan 4 (3-8 weeks after first treatment) in the left (2.8%, p=0.0043) and right hippocampus (4.3%, p=0.0001), right amygdala (4.3%, p=0.0002), and right thalamus (1.8%, p=0.0202) for the ECT group. However, only the right hippocampus and right amygdala passed a Bonferroni correction (p<0.0026). The TMS group showed a statistically significant volumetric increase in the right hippocampus (1.8%, p=0.0342) and right thalamus (1.5%, p=0.0482), but none of the areas passed the Bonferroni correction. As the perfusion has large normal variation, the small masks of the hippocampus, amygdala, and thalamus (HAT) were combined in each hemisphere for the perfusion analysis. There were no statistically significant changes between the participant groups at baseline perfusion, and none of the groups showed a statistically significant longitudinal change. There were also no statistically significant differences across the hemispheres. Although not significant, there were trends showing a longitudinal decrease in perfusion in the ECT group. Similarly, in the TMS group, trends showing a slight decrease followed by an increase in perfusion were observed. There were also trends showing an increased baseline perfusion for the ECT group compared to the other participant groups in both hemispheres. See Figure 1 and Figure 2 for volume and perfusion results, respectively. A volumetric increase in several of the selected anatomical structures was found for the patient groups despite the relatively low number of participants. In the ECT group, the left and right hippocampus, right amygdala and right thalamus showed a significant increase after treatment. The patients in the TMS group only had a significant increase in the right hippocampus and right thalamus, and the increases were smaller and less significant. This may indicate that the stronger ECT treatment will lead to a larger volumetric increase than the weaker TMS treatment.
The trend of a higher baseline perfusion for the ECT group in the left and right hemisphere, as well as the indicative changes in perfusion in the selected brain structures, will be interesting to explore in future analysis and larger data samples. MRI structural and perfusion images from healthy controls, ECT and TMS participants in an ongoing clinical study were analyzed. The study found a volumetric increase in selected subcortical brain structures after treatment, with more pronounced effects in the ECT treated participants than in the TMS group. These findings are in consistency with literature. Additionally, the study found trends in perfusion changes in these same structures which will be further explored in a larger patient sample.
Ingrid Kleive ANDERSEN (Bergen, Norway), Erling ANDERSEN, Frank RIEMER, Ute KESSLER, Leif OLTEDAL, Renate GRÜNER
13:30 - 15:00
#47146 - PG529 Multimodal MRI Biomarkers of Therapeutic Response in Depression Remission: Insights from Acceptance and Commitment Therapy.
PG529 Multimodal MRI Biomarkers of Therapeutic Response in Depression Remission: Insights from Acceptance and Commitment Therapy.
Depression remains a leading cause of suicide worldwide, contributing to over 700,000 deaths annually. Traditional antidepressant treatments often fail to address suicidal ideation, particularly in high-risk patients. Acceptance and Commitment Therapy (ACT), a third-wave cognitive behavioral therapy, has shown efficacy in reducing depression and suicidal ideation by targeting emotional regulation through mindfulness. Neurobiological models suggest that ACT may exert therapeutic effects by modulating brain networks such as the Default Mode Network (DMN) and the Salience Network (SN), which are implicated in rumination and emotional processing. This study aimed to identify MRI biomarkers of therapeutic response to ACT in adults with a history of suicide attempts.
We conducted a multimodal MRI study on adults undergoing either ACT or relaxation training (Relax), both in addition to standard treatment. MRI data were acquired before and after a 7-week intervention period. Data included structural MRI, diffusion tensor imaging (DTI), arterial spin labeling (ASL), and resting-state functional MRI (rs-fMRI). Using the Schaefer 400-parcel atlas, we extracted functional connectivity and cerebral blood flow (CBF) metrics, with a focus on DMN, SN, and ventral attention networks. A principal component analysis (PCA) was applied to clinical scores (depression, hopelessness, and psychological pain) to derive a single “negative component” representing symptom severity. Associations between imaging metrics and changes in this composite score were analyzed using linear models corrected for multiple comparisons (pFDR). A total of 87.5% of participants were female, with a mean age of 40 ± 12 years. At baseline, 81% were experiencing a major depressive episode. No significant group-by-session interaction effects were observed for anatomical or graph theory-based functional metrics. However, across both groups, clinical improvement (i.e., reduction of the negative component) was significantly associated with: (1) increased modularity within the Salience Ventral Attentional Network (r = –0.476, pFDR < 0.05), particularly in right medial cortical regions; (2) increased CBF in the right mid-cingulate cortex (r = –0.5, pFDR < 0.05); and (3) decreased functional connectivity between the left anterior cingulate cortex and the right superior frontal gyrus (T = 4.72, pFDR < 0.05). These findings support a neurobiological model in which therapeutic response in depression is linked to a reorganization of salience-related functional networks. Increased modularity within the Salience Ventral Attentional Network suggests a more functionally segregated and specialized brain organization following therapy. The observed changes in CBF and connectivity further highlight specific right medial brain regions as potential nodes of therapeutic plasticity. Improvement in depressive symptoms and suicidal ideation following ACT or relaxation training appears to be associated with functional reorganization within the Salience Ventral Attentional Network. This reconfiguration, marked by increased modularity and region-specific changes in CBF and connectivity, may enhance emotional and cognitive regulation, thereby supporting clinical remission.
Guillaume CLAIN (Montpellier), Jeremy DEVERDUN, Manon MALESTROIT, Olie EMILIE, Veronique BRAND-ARPON, Deborah DUCASSE, Philippe COURTET, Emmanuelle LE BARS
13:30 - 15:00
#47607 - PG530 Multimodal 7T MRI and Machine Learning for Stratifying ALS Progression in Small Cohorts.
PG530 Multimodal 7T MRI and Machine Learning for Stratifying ALS Progression in Small Cohorts.
Amyotrophic Lateral Sclerosis (ALS) is a rare, progressive neurodegenerative disorder with notable heterogeneity in clinical presentation and disease progression [1]. Accurate early diagnosis and prognosis are critical for effective patient management and treatment personalization. Current diagnostic methods largely depend on clinical evaluation, often leading to delays in diagnosis and prognostication [2]. Recent advances in ultra-high field (7T) MRI enable acquisition of structural, diffusion, and metabolic information at higher spatial resolution, potentially improving ALS characterization through imaging analysis [3,4]. Our study evaluates the use of such multimodal 7T MRI via suitable machine learning (ML) approaches to identify imaging biomarkers capable of differentiating healthy individuals from ALS patients, and further stratifying patients into slow and fast progressors.
The study enrolled 30 subjects, comprising of 16 ALS patients, and 14 healthy controls. We employed advanced 7T multimodal neuroimaging techniques, including structural, diffusion tensor imaging (DTI), and sodium imaging to obtain 270 MRI-based parametric samples. The parametric maps consisted of quantitative T1 (qT1), fractional anisotropy (FA), mean diffusivity (MD), axial diffusivity (AD) and radial diffusivity (RD), total sodium concentration (TSC), sodium T2*short (T2s) and T2*long (T2l) relaxation times, as well as the sodium signal fraction (fNa), representing the sodium signal from the short component [5]. The brain was parcellated into 332 regions of interest (ROIs), incorporating both gray matter (GM) and white matter (WM). Mean ROI values across the nine imaging parameters were extracted per subject, resulting in a high-dimensional feature space. The ML pipeline was developed after thoroughly evaluating various step configurations. A nested cross-validation framework was adopted using leave-one-out cross-validation (LOOCV) for robust performance estimation, and compared against chance-level predictions. Classification tasks included: (i) controls vs ALS patients (2-class), (ii) controls vs slow vs fast progressors (3-class), and (iii) slow vs fast ALS progressors. Based on our systematic analysis of ML pipeline design, a multinomial logistic regression (MLR) classifier with L2 regularization was selected, with standard scaling feature normalization and hyperparameter tuning. As seen in tables 1 and 2, unimodal analyses showed that among the imaging markers, TSC, MD and RD were most informative, particularly when WM ROIs were included in the analysis. Multimodal fusion helped improve the classification outcomes, with the 3-class setup achieving 70% accuracy and a Z-score of 5.2 when using the full GM+WM parcellation. Classification between slow and fast progressors improved from 69% (Z=1.74) to 88% (Z=3.96) when healthy controls were included during training. Fig. 1 illustrates the outcome of feature importance analysis, revealing that sodium imaging parameters contributed 60% of top selected features, followed by DTI-derived features (30–40%). Structural qT1 imaging provided limited discriminative value. The analysis highlighted both GM and WM ROIs, thereby emphasizing the need for whole-brain evaluation. This study demonstrates the feasibility and clinical utility of integrating multimodal 7T MRI with machine learning to classify ALS patients and predict disease progression in a small-cohort setting. White matter alterations, as captured by DTI metrics (RD, MD) and metabolic dysregulation indicated by 23Na imaging (TSC, fNa), emerged as dominant biomarkers. The inclusion of healthy controls during training enhanced the model's robustness in stratifying ALS progression, likely by providing contrast for learning disease-relevant patterns. Conventional structural imaging (qT1) showed limited value, underscoring the added value of advanced imaging modalities, thereby confirming the choices made for the ongoing multimodal dataset acquisition. The study also established that extensive pipeline optimization yielded only marginal improvements, emphasizing the greater impact of data enrichment strategies such as multimodal fusion. Our findings highlight the potential of combining advanced 7T MRI modalities with classical machine learning approaches to identify robust imaging biomarkers for ALS diagnosis and prognosis in small cohorts. Sodium imaging and DTI-derived features offer substantial discriminative power, especially when both gray and white matter brain regions are analyzed. Importantly, integrating healthy controls during training and leveraging multimodal MRI data enhances the model’s ability to stratify ALS patients by progression rate. This work lays a methodological foundation for future studies in rare neurodegenerative diseases, advocating for data enrichment and multimodal integration over complex pipeline tuning when dealing with limited sample sizes.
Shailesh APPUKUTTAN (Marseille), Aude-Marie GRAPPERON, Mounir Mohamed EL MENDILI, Hugo DARY, Maxime GUYE, Annie VERSCHUEREN, Jean-Philippe RANJEVA, Shahram ATTARIAN, Wafaa ZAARAOUI, Matthieu GILSON
13:30 - 15:00
#47916 - PG531 Brain resting-state fMRI signal complexity in temporal lobe epilepsy (TLE) patients.
PG531 Brain resting-state fMRI signal complexity in temporal lobe epilepsy (TLE) patients.
Temporal lobe epilepsy (TLE) is the most common form of focal epilepsy in adults, characterized by seizures originating in the temporal lobe [1]. Antiepileptic medications can effectively control symptoms in 70–80% of patients, but accurate diagnosis remains challenging [2]. Many conditions mimic epileptic seizures, and routine anatomical magnetic resonance imaging (MRI) and electroencephalography (EEG) often yield normal results in TLE patients due to subtle or absent structural abnormalities and the limited spatial resolution of EEG. Resting-state functional MRI (rs-fMRI) may be a promising alternative method for detecting TLE. This technique measures spontaneous, low-frequency fluctuations of the blood oxygen level–dependent (BOLD) signal, which indirectly reflects local neural activity while the subject is at rest, in the absence of a task/stimuli [3]. Complexity analysis, often quantified by the Hurst exponent, can be used to assess the temporal self-similarity and predictability of rs-fMRI signals [4]. This approach has shown utility in identifying and classifying certain neurological disorders, as reduced brain complexity has been consistently associated with disease [5,6]. Thus, the objective of this work was to evaluate whether a similar analytical approach would be of diagnostic value for patients with TLE.
High-resolution T1-weighted spoiled gradient echo 3D structural images and rs-fMRI data from 9 participants with TLE (22.1 ± 1.9 years old; 5 males and 4 females) and 10 age- and sex-matched healthy controls (22.3 ± 1.9 years old; 5 males and 5 females) were accessed from the Epilepsy Connectome Project (ECP) database [7]. Clinical and diagnostic features of the TLE group are summarized in Table 1. Rs-fMRI data were acquired on a 3T GE Discovery MR750 MRI with the following parameters: GE-EPI, 2mm isotropic, FOV=20.8cm, TE/TR=33.5/802ms, and 377 time points per run.
Rs-fMRI preprocessing steps were performed using FSL [8,9]. This included (1) discarding the first five functional volumes to allow for magnetization equilibrium, (2) eddy current and motion correction, (3) spatial smoothing with Gaussian kernel and FWHM=5mm, (4) 4D global normalization, (5) brain extraction, (6) registration to MNI standard space, and (7) high-pass temporal filtering at 0.01Hz.
The Hurst exponent was then computed voxel-wise in grey matter using rescaled range (R/S) analysis [4,10]. Group differences were evaluated using a linear mixed-effects model (3dLME in AFNI) to assess the effects of sex and disease status. Our analytical approach identified sizable regions of interest (ROIs) in the left superior temporal gyrus and the right hippocampus and amygdala (Fig. 1), where TLE patients showed reduced rs-fMRI signal complexity compared to healthy controls, indicated by a significantly higher Hurst exponent (Fig. 2). According to the ECP database [7], neither of these regions showed evidence of sclerosis or other abnormalities on standard clinical MRI, suggesting that the observed complexity reduction occurred in the absence of overt structural changes. We observed a reduction in rs-fMRI signal complexity in several temporal lobe regions in TLE patients. The healthy brain is best described as a complex system, and reduced rs-fMRI signal complexity may reflect a diminished capacity to adapt to dynamic demands [3]. In TLE, this loss of complexity could indicate disrupted or less flexible neural dynamics resulting from recurrent seizure activity or network reorganization [11]. Our findings support the idea that complexity analysis can capture functionally meaningful and spatially specific changes in the BOLD signal, in regions that appear normal on conventional anatomical imaging. While previous studies have reported reduced signal complexity in TLE using EEG, this is, to the authors’ knowledge, the first to examine these alterations using rs-fMRI, which offers superior spatial resolution for localizing brain activity. This approach may offer new insights into how epilepsy affects brain organization and communication. It could also potentially represent a novel biomarker for TLE diagnosis, especially in cases where structural imaging appears normal. Our next step will be to increase the sample size and apply subject-level analyses to evaluate whether this method can support personalized diagnosis.
Molly ARMSTRONG (Hamilton, Canada), Alejandro AMADOR-TEJADA, Michael NOSEWORTHY
13:30 - 15:00
#47767 - PG532 Quantifying and correcting axonal shrinkage in transmission electron microscopy: cryo-fixation vs. Epon-embedding.
PG532 Quantifying and correcting axonal shrinkage in transmission electron microscopy: cryo-fixation vs. Epon-embedding.
Axon diameter and g-ratio are key microstructural parameters modulating saltatory conduction velocity (CV) in white matter tracts [1]. However, different tissue processing methods and imaging modalities yield varying axonal diameters, leading to divergent interpretations of CV properties [2].
Diffusion MRI (dMRI) enables in vivo estimation of axon diameter but relies on biophysical models for summary statistics. Validation often uses transmission electron microscopy (TEM) or synchrotron X-ray nano-holotomography (XNH) [3-5]. Tissue preparation for these modalities often involves dehydration and resin embedding, causing significant but poorly understood shrinkage (0-65% [2]). In contrast, cryo-fixation uses high-pressure freezing (HPF) of fresh tissue to preserve cellular integrity and extra-axonal space, as shown in the cortex [6].
Here, we use deep-learning-based segmentation to quantify axon diameter and myelin thickness in the rat corpus callosum (CC) for HPF cryo-fixed and regular Epon-embedded tissue, quantifying and correcting the well-known tissue shrinkage effect in resin-embedded samples.
Eight young male Sprague-Dawley rats (NTac:SD-M; 4 weeks old) were used [2]. In each rat’s midsagittal slice, the CC midbody was sampled using a 1 mm biopsy punch. N=4 samples underwent cryo-fixation via HPF followed by freeze-substitution, and N=4 underwent standard resin embedding. TEM (Philips CM100) was performed at 4200x magnification, 11.9 nm isotropic pixel size, and ∼25 µm x 25 µm field of view. In total, 287 Epon and 299 cryo 2D TEM images were collected.
For segmentation, images were downsampled fourfold for computational efficiency. Axons and myelin were segmented with a U-Net, trained separately on 15 manually corrected images per modality with an expert-in-the-loop procedure. Initial segmentation used AxonDeepSeg [7]. Blob-based analysis isolated axons; inner diameter and myelin thickness were estimated from the minor axis of a fitted ellipse to account for eccentricity. Fig. 1 shows representative axon and myelin segmentations for cryo- and Epon-TEM images. Total histograms of axon diameter and myelin thickness modality-dependent differences are shown in Fig. 2. Larger axons, in the tail of the distribution (90th quantile), shrink more than the smaller axons near the mode. In contrast, myelin thickness histograms overlap, indicating no shrinkage.
The axon diameter Q-Q plot (Fig. 3a) reveals non-linear shrinkage in Epon-TEM: small axons are linearly underestimated up to the mode, whereafter underestimation of larger axons is non-linear. Axon diameters in Epon-TEM shrink by up to ∼50% relative to cryo-TEM, while myelin thickness shrinkage remains <11%. In contrast, myelin thickness (Fig. 3b) exhibits a near-perfect linear correspondence between cryo- and Epon-TEM, indicating minimal shrinkage.
Fig. 4 shows the application of a uniform 40% correction (commonly used in the literature [2]) vs. a non-linear correction derived from the 100-bin Q-Q analysis. The linear 40% correction cannot take into account the non-linear shrinkage effects observed. Our data confirm significant non-linear axonal shrinkage in Epon-embedded samples versus cryo-fixed tissue, assumed to be closer to the in vivo state. Cryo-TEM exhibited a broader tail in the axon diameter distribution, reflecting better preservation of large axons. Q-Q analysis revealed near-linear shrinkage near the mode and stronger non-linear underestimation farther from it, indicating diameter-dependent shrinkage as a result of the resin embedding sample preparation. Myelin thickness showed negligible shrinkage across modalities, consistent with its low water content and structural rigidity. Despite shrinkage, segmentation of small axons succeeded in both modalities, unlike dMRI, which cannot resolve fine structures due to hardware limits. Interpolating the Q-Q curve, we corrected Epon-TEM axon diameters, aligning them closer to cryo-TEM and dMRI estimates. However, conventional linear corrections (0-65%) do not ensure accurate correction for shrinkage and risks underestimating large axons. This may contribute to the widely known issue that dMRI-based axon diameter estimates tend to overestimate axon diameters, even after applying 40% linear shrinkage correction to Epon-TEM histograms and accounting for dMRI’s volume-weighted metrics [2-3]. To assess axon diameters, tissue-processing-induced shrinkage must be considered. Modality-specific non-linear corrections enhance quantitative accuracy and structure-function predictions. Larger axons, likely due to higher water content, are more prone to dehydration shrinkage during Epon embedding, while myelin, with its low water content, remains more stable. Our findings stress the importance of accounting for non-linear shrinkage in structural components for robust morphometric comparisons across modalities.
Miren Lur BARQUIN TORRE (Copenhagen, Denmark), Christian S. SKOVEN, Mariam ANDERSSON, Tim B. DYRBY
13:30 - 15:00
#47366 - PG533 White matter damage of the cbgtc loop in progressive supranuclear palsy.
PG533 White matter damage of the cbgtc loop in progressive supranuclear palsy.
Progressive Supranuclear Palsy (PSP) is a rare neurological disease caused by damage to nerve cells controlling body movements. Often misdiagnosed as Parkinson’s Disease (PD), PSP falls under the classification of atypical Parkinsonism, displaying various differences in symptoms and pathology. Previous MRI studies have revealed distinguishing signs that can identify this disease in patients. However, such signs only display after advanced disease progression, lending it little value for use in clinical prognosis. The present study sought to enhance the understanding of the progression of PSP though fixel-based analysis (FBA). By isolating unique tracts affected by the disease, the authors hope to provide further directions for research into early identifying markers of PSP before it progresses to an advanced stage.
Images from 23 patients diagnosed with PSP (8 men and 15 women; mean age: 67.8 ± 6.5 years) and 23 healthy controls (8 men and 15 women; mean age: 67.2 ± 5.9 years) were analyzed. Participants were recruited from the outpatient clinics of the Department of Neurology of a tertiary medical center. PSP presence was established by senior neurologists using the criteria outlined by Litvan et al. (4) and supported by nuclear medicine examinations (i.e. TRODAT). Severity was evaluated using the Unified Parkinson Disease Rating Scale (UPDRS), and disease staging was assessed by the Modified Hoehn and Yahr Staging (MHY) scale. Postural impairment and gait disorder staging (PIGD) was calculated based on their respective scores in the UPDRS. Patients with PSP were divided into 2 groups based on disease duration: long (>5 years, N=10) and short (≤5 years, N=13).
Diffusion-weighted images were acquired from a 3T MR scanner (Magnetom Trio; Siemens, Erlangen, Germany) using a spin-echo echo planar imaging (SE-EPI) sequence with these parameters: repetition/echo time = 5700/108 ms, voxel size=2×2×3 mm3, flip angle=90°, acquisition matrix=96×96, FOV=192×192 mm2, 40 slices to cover the brain above the cerebellum. 160 contiguous axial T1-weighted images were acquired with the magnetization-prepared rapid acquisition gradient echo (MPRAGE) sequence with these parameters: repetition/echo/inversion time = 2000/2.63/900 ms; flip angle=9°, acquisition matrix=224×256, FOV=224×256 mm2, with 1×1×1 mm3 resolution. FBA was conducted with MRtrix3 (version 0.3.15, www.mrtrix.org). Multi-tissue constrained spherical deconvolution was utilized to estimate the fiber orientation distribution function (FOD) of each voxel. The FOD was normalized across all participants using symmetric diffeomorphic non-linear transformation FOD-based registration with a study-specific template. Fixel-specific measures were computed from each voxel. Statistical analysis was performed with SPSS. Age differences were evaluated by the Student’s t-test with a significance level of p<0.05. Statistical analyses of images were done in MRtrix3, with both age and sex used as covariates. Connectivity-based fixel enhanced (CFE) and non-parametric permutation testing were used to evaluate the differences in fixel-based metrics, while linear regression between fixel-based metrics and clinical assessment scores (UPDRS-III, LEDD, and PIGD) was carried out with the general linear model. The statistical significance threshold was set at a FWE-corrected p < 0.05. Patients with PSP displayed a reduction in FD (fiber density), FC (fiber cross-section), and FDC (fiber density & cross-section) in the bilateral superior cerebellar peduncle, cerebral peduncle, posterior limb of internal capsule, and left corticospinal tract, and reductions in FD and FDC were seen in the bilateral posterior thalamic radiation, posterior corona radiata, retrolenticular part of internal capsule, and splenium and body of corpus callosum compared to controls. Compared to the short-term group, patients with a disease duration of >5 years displayed significant differences in UPDRS scores, as well as more widespread affected brain regions. Reductions in FD and FDC were observed in the corpus callosum, bilateral superior and posterior corona radiata, posterior limb of internal capsule, posterior thalamic radiation, and cingulum. A significant correlation was found between clinical parameters and fixel-based metrics in all patients compared to control. By isolating affected tracts, the findings of this study provide future directions for research into the identification and management of PSP. This study was constrained by a small sample size due to PSP's rarity. In addition, due to the asymptomatic nature of early-stage PSP, the patients from which PSP data was collected was limited to those who exhibited clear symptoms. Therefore, more longitudinal research is needed to collect data from patients during the early period of PSP. Through FBA, significant white matter tracts implicated in PSP were isolated, with significant differences observed between long-term and short-term groups.
Benjamin CEDERBERG (Taoyuan, Taiwan), Chih-Chien TSAI, Jiun-Jie WANG
13:30 - 15:00
#45750 - PG534 Alteration in hippocampal subfields and thalamic and amygdala nuclei volume and their association with cognition in mild cognitive impairment and glycemic state.
PG534 Alteration in hippocampal subfields and thalamic and amygdala nuclei volume and their association with cognition in mild cognitive impairment and glycemic state.
Alzheimer’s disease (AD) is the most prevalent neurodegenerative disorder in older adults. Mild cognitive impairment (MCI) is recognized as an intermediate stage between normal cognition and mild dementia, characterized by memory deficits(1). Hippocampus, amygdala and thalamus are responsible of cognitive functions. Moreover, recent studies suggest that there is a link between Alzheimer disease and hippocampal atrophy and insulin resistance(2,3).
To investigate this, we conducted magnetic resonance (MR) volumetry of healthy control and Mild Cognitive Impairment patients. Mini Mental State Exam (MMSE) was assessed and fasting glycemia and Hemoglobin A1C (HbA1c) were measured, and we conducted the correlation analysis between volumetry and theses parameters.
25 healthy control (22f/3m), age 70.1±6.1, 33 patients (30f/3m) diagnosed as MCI, age 70.1±7.3, participated the study. Mini Mental State Exam (MMSE), HbA1c and fasting glycemia were measured.
Study was performed by 3T MR system (PrismaFit) using a 64-channel head coil (Siemens Healthcare, Germany).
T1-weighted MPRAGE was acquired for the volumetry of the hippocampal and amygdala subfields and thalamus nuclei with the voxel size 0.83mm3. Freesurfer version 7.4.1 was used. Additional T2w-FLAIR was acquired and used for the segmentation of the hippocampal and amygdala subfields(4). Thalamus nuclei segmentation was conducted(5,6). We conducted the Mann-Whitney U test by SPSS (version 29.0.2.0) to see the difference in volumetry between two groups.
Spearman correlation analyses were performed using R (version 4.4.3) to assess associations between brain volumetric measures and cognitive/metabolic variables, including MMSE, fasting glucose, and HbA1c. Missing data were present across variables: MMSE (2 HC, 2 MCI), fasting glucose (3 HC, 2 MCI), and HbA1c (5 HC, 4 MCI); analyses were conducted using pairwise deletion. The Mann-Whitney U test revealed significant volumetric reductions in Left Basal nucleus, left Accessory Basal nucleus, Left Anterior amygdaloid, Left Central nucleus, Left whole amygdala, Right Basal nucleus, Right Accessory Basal nucleus, Right Anterior amygdaloid, Right Central nucleus, Right paralaminar nucleus and Right whole amygdala from Amygdala nuclei in MCI group. Regarding the Hippocampal subfields, Left presubiculum head, Left CA1 head, Left GCMLDG head, Left CA3 head, Left CA3 head, Left whole hippocampal head, Right hippocampal fissure, Right presubiculum head and Right whole hippocampal head showed significant reductions in MCI group. Regarding the thalamic nuclei, Right LD showed significant decrease.
The results of the correlation analysis are displayed in Figure (A),(B), and (C). Significant reduced volumes in both sides of the amygdala and hippocampus were reported in MCI group, on the other hand there was no significant reduction in thalamic structures. It is a contradiction to one study stating that the early loss of the thalamic volume in the MCI group is the early sign of the disease(7).
Several Amygdala nuclei and Hippocampal subfields structures show mild to weak correlations to MMSE, while glucose metabolic markers only showed a few correlations. Recent study suggested the reductions of right side of Amygdala nuclei are hallmark of the MCI(8). Regarding the Hippocampal subfields, one review says that there is a heterogenicity of the structural difference between MCI and control group, and from our study, bilateral presubiculum corresponds to the review consensus(9).
Regarding the thalamus, structures in right thalamus were negatively correlated to the level of HbA1C and fasting glucose although the structures on the left thalamus did not show the significant correlation. This could be related the dominance of the bran and the difference of the blood flow, as well as the lateralization preference of the formation of atherosclerosis due to the higher glycemia level. Significant reductions of the volumes in the amygdala nuclei and hippocampal subfields were observed in MCI group while no reductions were observed in thalamus structures in MCI group. Amygdala and Hippocampal structures showed mild to weak correlations to cognitive functions although there are not many correlations between these structures and glycemic markers. On the other hand, Thalamic nuclei in the right side showed mild to weak negative correlations to HbA1C and fasting glucose, while only a few correlations were observed between MMSE and the volumetry of the thalamic structures.
Kanji CHO (Vienna, Austria), Radka KLEPOCHOVA, Barbara UKROPCOVA, Josef UKROPEC, Lucia SLOBODOVA, Martin KRSSAK, Ivica JUST, Florian FISCHMEISTER
13:30 - 15:00
#45944 - PG535 Postmortem in-situ MRI of the medulla oblongata in amyotrophic lateral sclerosis: Assessing relaxation and DTI parameters.
PG535 Postmortem in-situ MRI of the medulla oblongata in amyotrophic lateral sclerosis: Assessing relaxation and DTI parameters.
Amyotrophic lateral sclerosis (ALS) is a clinically heterogeneous, fatal neurodegenerative disease. Its vast heterogeneity poses a challenge to the development of reliable disease-modifying therapies, which remain unavailable [1]. Sensitive biomarkers are urgently needed to enable successful clinical trials [2].
Although brainstem pathology is a hallmark feature of ALS [3], it has not yet been extensively investigated in imaging studies [4]. This study aimed to explore potential MRI biomarkers in the brainstem, particularly in the medulla oblongata, by assessing relaxation and DTI parameters. Postmortem (PM) in-situ MRI enables the assessment of end-stage ALS without fixation-related alterations seen in ex-situ measurements [5]. Another advantage is the possibility of direct comparison with histological data for validation.
All procedures were approved by the local ethics committee. PM in-situ (brain not extracted from the skull) whole-brain MRI scans were performed on six deceased male patients with clinically definite ALS, as defined by the revised El Escorial criteria [6], and on eight deceased individuals (five males, three females) with no known neurological conditions, who served as healthy controls (HC). The ALS and the HC groups were matched for age (ALS: 63.2 ± 8.2, HC: 69.4 ± 17.9. p = 0.30) and postmortem interval (ALS: 46.75 ± 20.0, HC: 28.3 ± 19.5. p = 0.14). Prior to the scan, the deceased subject was placed in a cooling chamber at 4 °C. Based on a temperature correction model developed in another study [see abstract #45942], the MD and T1 parameters were adjusted to a reference temperature of 20 °C to facilitate comparison between ALS and HC.
The scans were performed at 3T (Siemens MAGNETOM Prisma), including the following sequences:
• MP2RAGE [7]: 176 slices per slab, FoV = 240 × 256 mm, TR = 5000 ms, TE = 2.98 ms, TI = 700 and 2500 ms, flip angle = 4° and 5°, GRAPPA acceleration factor 3, isotropic resolution of 1 mm3. Used for masking and T1 maps.
• DTI: b = 2000 s/mm2, 64 isotropically distributed diffusion directions, 3 b = 0 s/mm2, TE = 109 ms, TR = 18700 ms, 100 slices, isotropic resolution of 1.8 mm3. Used for FA and MD maps.
• Multi-contrast spin-echo: 12 TEs, TR = 5720 ms, 44 slices, slice thickness 4 mm, in-plane resolution 1x1 mm2. Used for T2 maps. Sequence only performed for four ALS cases, but all HC cases.
• Multi-echo gradient-echo: 12 TEs, TR = 68 ms, flip angle 20°, 44 slices, slice thickness 4 mm, in-plane resolution 1×1 mm2. Used for T2* maps. Sequence only performed for five ALS cases, but all HC cases.
The masks for the medulla oblongata were manually created using 3D Slicer [8]. After correcting for eddy currents, FSL’s dtifit command was used to generate FA and MD maps. T1 maps were computed using a publicly available python script [9], while T2 and T2* maps were generated using a voxel-wise two-parameter mono-exponential single decay fit.
Comparisons between ALS and HC were conducted using the Mann-Whitney U test and visualised with box plots (using python). Due to the limited sample size, no significance level was defined. The comparison of relaxometry and DTI parameters between six ALS patients and eight HC suggests increased T1 values in the medulla oblongata of patients with ALS (see Figure 1). While median values for FA, MD, and T2 differed between groups, the corresponding p-values do not support clear differences between ALS and HC. The objective of this PM in-situ study was to explore potential MRI biomarkers in the medulla oblongata by comparing relaxation and DTI parameters between six patients with ALS and eight HC. The ALS group showed potentially increased T1 in the medulla oblongata. Visual inspection further suggested lower FA (p = 0.08) and higher MD values (p = 0.11) in patients with ALS. Increased T1 and MD, along with reduced FA, has been shown to indicate reduced myelin density [10]. However, the small sample size in the current study limits the ability to draw definitive conclusions. Further validation in larger cohorts, ideally incorporating histological confirmation as the ground truth, is necessary to support this hypothesis. The exploratory findings of this study could aid in the development of novel MRI biomarkers for ALS, which is a crucial prerequisite for successful clinical trials. However, studying larger cohorts along with corresponding histological validation is required to confirm the current findings.
Dominique NEUHAUS, Dominique NEUHAUS (Basel, Switzerland), Maria Janina WENDEBOURG, Eva SCHEURER, Regina SCHLAEGER, Claudia LENZ
13:30 - 15:00
#47356 - PG536 Peripheral Metabolite 3-Oxooctadecanoic Acid Correlates with Brain Microstructural Changes and Cognitive Impairment in End-Stage Renal Disease.
PG536 Peripheral Metabolite 3-Oxooctadecanoic Acid Correlates with Brain Microstructural Changes and Cognitive Impairment in End-Stage Renal Disease.
End-stage renal disease (ESRD) not only imposes systemic metabolic stress but also contributes to central nervous system dysfunction, particularly cognitive impairment. Previous studies have identified metabolomic alterations related to inflammatory and neurochemical changes in ESRD, such as elevated indoxyl sulfate [1], increased lipid metabolites [2], and altered phosphatidylcholine levels [3]. However, the role of peripheral blood metabolites in mediating cognitive dysfunction remains unclear. This study investigates the relationship between specific serum metabolites and brain microstructural changes associated with cognition in ESRD patients, using advanced metabolomic profiling and neuroimaging techniques.
Thirty-seven participants were enrolled in the study, including 14 healthy controls (mean age: 60.1 ± 10.7 years; 10 men), ESRD patients with normal cognition (mean age: 67.0 ± 6.5 years; 6 men), and ESRD patients with cognitive impairment (mean age: 73.5 ± 9.9 years; 6 men). MRI acquisition was performed using a 1.5T scanner and included T1-weighted magnetization-prepared rapid acquisition gradient echo sequences and diffusion-weighted imaging. Serum samples were subjected to untargeted metabolomic profiling using SYNAPT G2-MALDI mass spectrometry, identifying PC(18:2(9Z,12Z)/22:6(5Z,7Z,10Z,13Z,16Z,19Z)-OH(4)), 3-Oxooctadecanoic acid, and 10-alpha-methoxy-9,10-dihydrolysergol, which related to lipid metabolites and phosphatidylcholine levels alteration. Diffusion tensors were reconstructed using the Diffusion Kurtosis Estimator, generating tensor-derived metrics including mean diffusivity, radial diffusivity, axial diffusivity, and fractional anisotropy. Correlation analyses were conducted between metabolite concentrations and DTI metrics. A p-value of less than 0.05 was considered statistically significant, with Bonferroni correction applied for multiple comparisons. Among the three metabolites—PC(18:2(9Z,12Z)/22:6(5Z,7Z,10Z,13Z,16Z,19Z)-OH(4)), 3-oxooctadecanoic acid, and 10-alpha-methoxy-9,10-dihydrolysergol—only 3-oxooctadecanoic acid exhibited a statistically significant difference across the three groups (Figure 1). In addition, 3-oxooctadecanoic acid exhibited a consistent and robust correlation with diffusion tensor imaging metrics across multiple brain regions implicated in cognitive function. Specifically, elevated serum levels of 3-oxooctadecanoic acid were associated with increased mean diffusivity, axial diffusivity, and radial diffusivity in frontal, insular, and temporal areas. For mean diffusivity, significant correlations were observed in the middle frontal gyrus (MFG_A46, r = 0.579), inferior frontal gyrus (IFG_IFS, r = 0.601), insular gyrus (INS_dId, r = 0.577–0.588), and inferior temporal gyrus (ITG_A20cv and ITG_A20il, r = 0.599) (Figure 2). Similar trends were found for axial diffusivity in the middle frontal gyrus (r = 0.583–0.585), inferior frontal gyrus (r = 0.599), and inferior temporal gyrus (r = 0.585–0.637) (Figure 3). Radial diffusivity also showed significant correlations in the inferior frontal gyrus (r = 0.603), inferior temporal gyrus (r = 0.599), and insular cortex (r = 0.591) (Figure 4). This study identifies 3-oxooctadecanoic acid as a potential peripheral biomarker associated with microstructural brain alterations in patients with end-stage renal disease. Significant correlations were observed between serum levels of this metabolite and diffusion tensor imaging metrics in brain regions integral to cognition. These regions include the middle frontal gyrus and inferior frontal gyrus, which are involved in executive function; the insular cortex, associated with interoception and emotional regulation; and the inferior temporal gyrus, a key area for semantic memory and visual recognition. The positive correlations between higher 3-oxooctadecanoic acid levels and increased diffusivity suggest potential axonal degeneration or demyelination processes, which align with known cognitive and structural changes observed in ESRD. The metabolite’s consistent association with multiple DTI parameters across these regions supports its role as a marker of widespread white matter integrity loss. Additionally, the gradation in these effects across groups stratified by cognitive status further underscores the clinical relevance of 3-oxooctadecanoic acid in neurocognitive outcomes among ESRD patients. By integrating untargeted blood metabolomics with advanced neuroimaging, this study provides novel evidence linking elevated 3-oxooctadecanoic acid levels to region-specific brain microstructural abnormalities related to cognitive function in ESRD. These findings highlight the potential of peripheral metabolites as non-invasive biomarkers for early detection and monitoring of cognitive impairment in this high-risk population. Moreover, targeting the implicated metabolic pathways may offer new therapeutic strategies aimed at protecting brain health in patients with ESRD.
Chih-Chien TSAI, Yi-Chou HOU, Ruei-Ming CHEN, Jiun-Jie WANG (TaoYuan, Taiwan)
13:30 - 15:00
#47557 - PG537 Substantia nigra and posterior putamen MRI asymmetries reveal distinct pathology and explain motor symptom lateralization in Parkinson’s disease.
PG537 Substantia nigra and posterior putamen MRI asymmetries reveal distinct pathology and explain motor symptom lateralization in Parkinson’s disease.
MRI-based markers in the Substantia Nigra (SN) and Posterior Putamen (PP) offer promise for linking structural brain changes to motor dysfunction in Parkinson’s Disease (PD). Motor symptom asymmetry is a hallmark of early PD. Currently, the relationship between the different brain structures asymmetry, and their distinct contributions to symptom asymmetry, remains incompletely understood. Prior studies have used the ratio between T1-weighted (T1w) and T2-weighted (T2w) images to assess PP asymmetry while mitigating scanner variability [1]. Here, we demonstrate that when properly harmonized across visits and sites, T2w – and in combination, T1w and T2w – yield stronger and more interpretable associations of the SN and PP asymmetry with motor symptoms.
We analyzed data from 157 PD patients from the Parkinson’s Progression Markers Initiative (PPMI), totaling 402 MRI sessions. Each session included T1w, T2w, and proton density-weighted (PDw) scans acquired on 3T Siemens Trio TIM systems across 8 imaging centers. MRI preprocessing involved bias field correction and normalization using 3D polynomial fitting within white matter. SN was segmented using nonlinear transformation of the MASSP atlas [2]. The putamen was segmented using the FSL FIRST [3], and the PP was specifically delineated using mrGrad (https://github.com/MezerLab/mrGrad), as previously described [1].
Asymmetry indices (asym) were computed as left-right differences for each structure and modality. Motor asymmetry was assessed using MDS-UPDRS Part III (UPDRS3_asym) scores. We tested relationships between UPDRS3_asym and MRI-derived_asym using linear regression.
Control analyses included volumetrics, field bias analyses, and alternative regions (e.g., thalamus), to rule out global or artifactual effects. DaTSCAN data were included to compare MRI-derived asymmetries with dopaminergic deficits and assess MRI’s added explanatory value. All results were replicated within single visits. Harmonization substantially reduced inter-visit and inter-subject variability, enabling direct comparison of image intensities across visits and sites (Fig. 1).
Consistent with prior work [1], T1w/T2w_asym in the PP was correlated with UPDRS3_asym (r = 0.47, R² = 0.22, p < 10-23). Here, we also tested this association in the SN, which showed a significant correlation as well (r = -0.41, R² = 0.17, p < 10-18). We then extended the analysis by examining T1w, T2w, and PDw images separately. These separate analyses revealed that T2w_asym alone accounts for most of the observed associations, offering slightly stronger and more interpretable results than the T1w/T2w ratio. Specifically, T2w_asym in SN and PP were associated with UPDR3_asym, in opposite directions: SN positively (r = 0.42, R² = 0.18, p < 10-19; i.e., lower T2w contralateral to more affected side; Fig. 2a), and PP negatively (r = -0.47, R² = 0.22, p < 10-23; i.e., higher T2w contralateral; Fig. 2b). A combined SN–PP contrast (SN_asym − PP_asym) yielded a stronger association (r = 0.56, R² = 0.31, p < 10-34; Fig. 2c).
Multiple analytical approaches, including regression, PCA, and log-ratio transformations, converged on the same SN–PP contrast structure. PDw_asym yielded similar but weaker and oppositely signed effects, compared to T2w, in the SN and PP.
T1w_asym showed a significant effect in the PP but not the SN. Importantly, combining both T1w and T2w asymmetries in the PP using multiple regression outperformed their ratio (R² = 0.26, p < 10-26), suggesting that T1w and T2w carry additional information over T1w/T2w. A multimodal model including SN_T2w, SN_volume, PP_T2w, and PP_T1w explained the most variance in UPDRS3_asym (R² = 0.37, p < 10-38; Fig. 3).
Results were robust in single-visit analyses and remained significant beyond volumetric effects. Interestingly, MRI asymmetries measures explained unique variance beyond DaTSCAN, indicating complementary independent value. Findings support distinct pathological sources: in SN, T2w hypointensity likely reflects iron deposition; in PP, T2w hyperintensity and T1w hypointensity may reflect gliosis, atrophy, or microstructural disorganization.
Robust harmonization of the raw T1w and T2w signals, outperformed traditional T1w/T2w ratios. The SN–PP contrast offers a biologically grounded and cumulative marker of lateralized pathology.
Effects were specific to PD-relevant regions, robust across methods, and not explained by global asymmetry or preprocessing artifacts. Significant associations beyond DaTSCAN and volumetrics position multi-contrast MRI as a unique structural biomarker in PD. A contrast-based, region- and modality-specific MRI asymmetry model, focused on SN and PP, offers a robust and biologically interpretable marker of motor symptom lateralization in PD. Harmonized T1w, T2w, and PDw imaging captures complementary pathology beyond dopaminergic or volumetric measures.
Elior DRORI (Jerusalem, Israel), Aviv A MEZER
13:30 - 15:00
#47622 - PG538 Adapting a social reciprocity task for 7T-fMRI for the purpose of autism research.
PG538 Adapting a social reciprocity task for 7T-fMRI for the purpose of autism research.
Differences in social-emotional reciprocity are a hallmark of autism. The DSM-5 defines these differences as ranging from atypical social approach and conversational exchange, to reduced sharing of interests, emotions, or affect [1]. To assess these behaviors, the Interactive Drawing Task (IDT) was developed as a language-free measure of social reciprocity. Drawing does not require developed language skills or a high IQ and is therefore feasible in a wide range of individuals. The IDT has consistently revealed reduced reciprocal behavior in both autistic children and adults [2,3]. However, the neural substrates supporting the behaviors tied to social reciprocity remain unclear. In this study, we have adapted a digital version of the IDT (DIDT) for use in an MRI environment.
Participants (N=6, 2 women, mean age = 35 ± 9 years) underwent scanning on a 7T Philips Achieva MRI scanner using an 8Tx/32Rx head coil. During the 8.5-minute DIDT functional run, the participant and an examiner took turns contributing to a joint drawing using an MRI-compatible touchpad and pen. An MRI-compatible button box, operated with the left hand, allowed participants to initiate or suppress drawing input. The drawing task was implemented using a custom MATLAB code. Turn-switching was initiated by clicking the “Switch Turn” panel at the bottom of the screen (figure 1G), with the system recording drawing onset, durations, and a screen capture. Prior to the drawing task inside the MRI, participants had a practice session in the mock scanner to familiarise with drawing in this setting. For practise, participants were shown a screen with strings of letters or numbers to trace over (figure 1A-F). Participants selected a pen color (purple or orange), while the examiner always drew in black.
Functional data were acquired using a 3D-EPI sequence (1.8 mm isotropic; TR/TE/TA = 44ms/17ms/1.37s; flip angle = 13°; SENSEyz = 2.6×3.27; FOV = 200×200×176mm³; 98 sagittal slices), along with a top-up scan for distortion correction. For coregistration purposes, high-resolution anatomical images were acquired using MP2RAGE (0.55 mm isotropic; TI1/TI2 = 1000/3000 ms; TR/TE = 6.3/2.9 ms; flip angles = 8°/8°; SENSEyz = 2.3×2.1).
Participant turn durations and drawing onset times were modeled as regressors in individual-level GLMs, followed by group-level analysis using FEAT. Specifically, we examined BOLD responses during participant drawing turns versus passive observation of the examiner’s drawing, and separately compared drawing onset to turn-switch events (onset > turn-switch). Motion correction was performed with MCFLIRT. 3D activation maps were normalized to MNI space; cerebellar activity was projected to a SUIT flatmap [4,5]. The DIDT was well-tolerated and rated as enjoyable by all but one participant, who reported a neutral experience. Data from one participant were excluded due to excessive head motion, the remaining five participants were included in the analysis. On average, participants completed 11.6 ± 3.91 drawing turns, with 11 ± 4.32 handovers back to the examiner. The average duration of each turn was 26.02 ± 8.72 s for participants and 16.97 ± 4.36 s for the examiner. Head motion during the fMRI runs was generally low across included participants (mean displacement: 0.93 ± 0.5 mm), supporting the task’s feasibility in most cases.
Significant BOLD responses during participants’ drawing turns were observed in primary motor and somatosensory cortices, as expected (Figure 2A). Additionally, the anterior insula was engaged bilaterally (Figure 2B). In contrast, passive observation of the examiner’s drawing yielded responses in the visual cortex (Figure 2A), which was expected, as well as in the temporo-parietal junction (TPJ), and cingulate gyrus (Figure 2C)—regions implicated in social cognition and mentalizing. Bold response in the putamen was observed right at the drawing onset (Figure 2D).
The BOLD response in the cerebellum (Figure 3) extended beyond the motor regions involved in right-hand movement towards areas implicated in working memory, divided attention, and emotional or narrative processing as identified with a multi-domain task battery [5]. The DIDT robustly engages cortical and cerebellar regions implicated in social-affective cognition. BOLD response in the insula, cingulate, TPJ, and cognitive cerebellar zones indicates that this nonverbal collaborative task successfully engages a broad social-affective neural network [5–8]. While one participant's data was excluded due to motion artifacts, the task was successfully completed and tolerated by the remaining participants, supporting its general feasibility in the high-field MRI scanner environment. This pilot study supports the DIDT’s potential as a nonverbal, high-field fMRI tool for mapping reciprocity-related processes. Future work in autistic populations will help elucidate how individual differences in social cognition are reflected in cortical and cerebellar engagement.
Amina ZIDANE BURGESS (Amsterdam, The Netherlands), Dylan VAN DER WAAL, Ryan MUETZEL, Sander BEGEER, Tineke BACKER VAN OMMEREN, Wietske VAN DER ZWAAG
13:30 - 15:00
#47710 - PG539 Brain network reorganization after adapted cognitive-behavioral therapy and immersive virtual reality in autism spectrum disorder with intellectual disability.
PG539 Brain network reorganization after adapted cognitive-behavioral therapy and immersive virtual reality in autism spectrum disorder with intellectual disability.
Individuals with autism spectrum disorder (ASD) and comorbid intellectual disability (ID) often show severe social anxiety (1), linked to atypical interactions among key brain networks: default mode (DMN), salience (SN), and dorsal attention (DAN) (2-4). Effective cognitive-behavioral therapy (CBT) in this population requires specific adaptations. This pilot study evaluates a novel intervention combining adapted CBT (ACBT) with immersive virtual reality (IVR) to improve social anxiety (5-7), examining changes in resting-state functional connectivity (rsFC).
Ten participants with ASD, mild ID and social anxiety (DSM-V-TR) were assessed at baseline and post-intervention. Combined ACBT-IRV intervention program spanned 12 weeks, comprising 24 sessions evenly divided into 6 weeks of individual therapy and 6 weeks of group therapy, including four blocks: Psycho-education, Social Skills Training, Behavioral/Cognitive Restructuring, and Systematic Desensitization/Exposure. A three-month follow-up was conducted to assess outcomes. At baseline and post-intervention sessions, social anxiety (Social Phobia Inventory), self-esteem (Rosenberg), and cognitive variables, including attention (d2) and planning (Tower of London) were evaluated using standardized tests, and functional data was acquired using a 3T MRI scanner
rsFC analysis focused on ROI-to-ROI correlations between DMN, DAN, SN nodes, and bilateral hippocampi/amygdalae (HCP-ICA atlas). Preprocessing and ROI-to-ROI analyses used the CONN toolbox. Between session effects (baseline - post-intervention) ROI-to-ROI rsFC changes were assessed using paired t-tests. Graph-theoretical metrics were computed on a subnetwork of selected ROIs (edges with z-score > 2.5). Associations between rsFC changes and emotional and cognitive changes were tested using linear models with delta-change scores. Secondary models examined baseline connectivity's predictive value for behavioral outcomes. Significance was set at p < 0.05, uncorrected due to the exploratory nature. After ACBT-IVR intervention, the left intraparietal sulcus (IPS, DAN) showed increased rsFC with SN nodes and decreased rsFC with DMN and hippocampal regions. Connectivity within the hippocampi–amygdalae cluster also increased. In addition, graph-theory analysis revealed increased global efficiency, degree, and betweenness centrality for the left IPS, suggesting a more central network role in the ASD/ID participants. Conversely, DMN nodes (medial prefrontal and lateral parietal cortices) became more functionally segregated, showing higher eccentricity and lower closeness centrality.
rsFC changes correlated with cognitive improvements (p < 0.01). Improved planning, assessed via the Tower of London test, was inversely associated with rsFC changes between DAN and SN, and between DMN and DAN. Increased perceived precision in the attention test d2 correlated with enhanced rsFC between lateral parietal cortices, DMN, and left amygdala nodes. Self-esteem improvements showed a negative association with internal SN rsFC. Baseline interhemispheric hippocampal rsFC strongly predicted social anxiety reduction (p < 0.001, R² = 0.85), with higher baseline connectivity linked to greater symptom improvement. Including post-intervention rsFC changes improved model fit (R² = 0.88). Only the bilateral hippocampal connection exhibited both significant post-intervention change (p < 0.01) and association with behavioral outcomes. Results suggest that the ACBT-IVR intervention is associated with rsFC reorganization in participants with ASD/ID. The dorsal attention network (DAN) became more integrated, while the default mode network (DMN) became more segregated, potentially reflecting enhanced external attention and reduced self-referential processing, consistent with neural correlates of reduced social anxiety (6,8). The left intraparietal sulcus (IPS) emerged as a key hub, and cross-network decoupling (e.g., DMN–DAN) was linked to improved cognitive performance, suggesting reduced attentional interference enhances executive function. Baseline interhemispheric hippocampal rsFC strongly predicted social anxiety reduction, indicating its potential as a biomarker of intervention effectiveness. This pilot study provides preliminary evidence that ACBT-IVR modulates rsFC in key social cognition networks in ASD/ID. Changes in DMN–limbic connectivity may reflect improved self-referential and emotional processing (9,10). A targeted intervention combining ACBT and IRV drives adaptive brain network reorganization in individuals with ASD and mild ID. By enhancing attentional network integration and reducing default mode network interference, this approach yields significant cognitive and emotional improvements. Baseline hippocampal connectivity emerges as a promising biomarker for predicting therapeutic success. These compelling findings pave the way for transformative interventions and call for rigorous validation in larger, controlled trials.
Elena DE LA CALLE (Girona, Spain), Melissa SAMANIEGO-REINOSO, Carles BIARNÉS, Oren CONTRERAS-RODRÍGUEZ, Victor PINEDA, Laura VERGÉS, Susanna ESTEBA-CASTILLO
13:30 - 15:00
#47781 - PG540 Diffusion Tensor Imaging assessment of offspring brain neurodevelopment in mouse model of diet-driven Autism Spectrum Disorder.
PG540 Diffusion Tensor Imaging assessment of offspring brain neurodevelopment in mouse model of diet-driven Autism Spectrum Disorder.
Clinical and preclinical studies, highlight the impact of maternal obesity and exposure to a high-fat diet (HFD) during pregnancy and lactation on an increased risk of symptoms of autism spectrum disorder (ASD) in the offspring. However, little is known about the processes by which an inappropriate environment of intrauterine and early childhood development interferes with normal development and brain function in offspring.
The aim of this work was to use magnetic resonance diffusion tensor imaging (DTI) and a mouse model of diet driven ASD, to find quantitative indicators of alterations in offspring’s brain structure influenced by mother exposure to HFD.
Obesity in C57BL/6J mice was induced by administering a high-fat diet (HFD) containing 45% energy from fat, while the control group received a standard control diet (CD) with 10% fat. After 8 weeks on the diet, females were mated to males and then kept on CD or HFD during pregnancy and lactation. After weaning, the offspring were kept on a standard diet for the remainder of the study. Male animals at the age of 58 weeks were used in this study. The number of the CD and HFD offspring was 9 per group.
After anesthesia, the animals were subjected to intracardiac perfusion with ice-cold phosphate-buffered saline (PBS) followed by 4% paraformaldehyde (PFA). The perfused brains were dissected immediately after perfusion and placed in an icy 4% PFA overnight, then the brains were rinsed three times in PBS. The brains were cryoprotectively fixed in an increasing gradient of 10-30% sucrose solution.
DTI imaging was performed on an MR 9.4T scanner, (Bruker BioSpin MRI GmbH), using DtiStandard SpinEcho pulse sequence with the settings: TE/TR of 20.3/5000 ms and / of 10/5 ms. A DTI scheme with 30 diffusion sampling directions at a b-value of 2297.5 s/mm² was used to obtain images with an in plane resolution/layer thickness of 0.1/0.2 mm, scaled to an 3D isotropic resolution of 0.1 mm.
DSI Studio (http://dsi-studio.labsolver.org/) was used to calculate diffusion anisotropy indexes and perform tractographic analysis. Fig. 1 shows examples of cross sections through DTI scans for control and high fat diet brains. Fig.2 shows comparison of fractional anisotropy values for brain structures where statistically significant differences between control and high fat diet brains were found. Fractional Anisotropy: The result of the intergroup difference in 6 out of 9 structures examined was statistically significant (p < 0.05), which is consistent with the observations in human infants, as know from scientific literature. After birth, general brain hypertrophy is observed at the early childhood stage, which is confirmed by magnetic resonance imaging in cohort of longitudinally examined infants aged 6 to 24 months (Shen, M. D., et al. 2013).
Tractography: Only for three structures did the results of intergroup differences come out statistically significant. In the case of the entire brain structure, the HFD group showed a higher result of the degree of elongation of nerve fibers (axonal packages) and a lower value of the diameter of nerve tracts. Statistically significant differences in values of Fractional Anisotropy were found between offspring’s brains, depending on the mother’s diet during pregnancy.
Artur RYŚ, Krzysztof JASIŃSKI, Katarzyna KALITA, Dawid GAWLIŃSKI, Władysław WĘGLARZ (Kraków, Poland)
13:30 - 15:00
#47810 - PG541 Quantitative analysis of the neuromelanin signal for differentiation In ams patients vs controls: a multiparametric approach.
PG541 Quantitative analysis of the neuromelanin signal for differentiation In ams patients vs controls: a multiparametric approach.
Multisystem atrophy (MSA) is a neurodegenerative disease that is often confused with Parkinson's disease (PD) due to similar clinical symptoms, complicating diagnosis and delaying patient management. Recent advances have identified neuromelanin (NM) as a potential biomarker for PD [1]. Our research therefore focuses on studying the NM signal between patients with AMS (N=38) compared with control subjects (SC) (N=20) in order to validate a biomarker for AMS.
Multicentre MRI and Dat Scan protocol (9 french reference centres)
1.Acquisition and mapping:
-Sequence dedicated to NM: Optimised T1 spin echo (TR = 900ms; TE = 10ms; 0.7x0.7x1.4).
-T1 and T2* gradient echo (TR= 50ms; ∆TE = 5 :5 :35ms; DFA= 5° - 20°; 0.7x0.7x1.4). These sequences were used to generate T1 and T2* maps with the hMRI toolbox [2].
-SPECT sequence with low-dose CT (110mA, 120 kV, slice thickness 3.75mm and a pitch of 1.1)
2.Segmentation
MRI: Sisyphe software was used for bi-operator segmentation of the NM. A logical And operation was then used to extract the area common to the segmentations. The Dice coefficients, 0.76 for control subjects and 0.70 for AMS patients, confirmed a good match between the segmentations.
Dat Scan: Segmentation of the striatum and analysis of volumes using DaTsoft3D software, developed by Pierre Gantet [4]. Use of DaTsoft3D software [4] to quantify the density of dopamine transporters (DaT) in the striatum and measurement of a binding potential (BP) resulting in a ratio of activity concentration between the striatum and a non-specific zone.
3.Extraction of radiomics
Use of 3D Slicer for radiomics extraction. These are extracted from the optimised T1 NM spin echo, R1 and R2* at AMS and SC. Two types of radiomics are studied here: shape radiomics and first-order radiomics. Shape radiomics provide volume information. First-order radiomics, on the other hand, are based on the histogram, such as the mean, variance, energy and entropy, indicating local variations in intensity within the ROI.
4.Data analysis
To validate the radiomics, two-tailed T-tests were performed. A P value < 0.05 indicates significance between groups. NM signal: No significant difference observed in T1 signal intensities or R1 values between groups, suggesting preservation of the longitudinal relaxation properties of NM. Significant difference (p = 0.0009) observed in the median R2* values.
NM radiomics: Radiomic analysis revealed characteristics that distinguish our two groups, such as energy (p=0.01) and entropy (p=0.009).
NM volume: A 23% decrease in absolute NM volume was measured (p<0.001) in AMS patients compared with SC patients. This observation was also made on Dat Scan images (p<0.001). Furthermore, the binding potential in the striatum was significantly reduced in AMS compared with SC (p<0.001). NM signal: The difference in the R2 signal indicates an alteration in the transverse relaxation properties, possibly linked to changes in the concentration of paramagnetic metals within the NM in AMS patients.
NM radiomics: The resulting significant radiomics converge towards a single interpretation. In SCs, the NM appears to be a complex, detailed and heterogeneous structure. In contrast, in AMS, the NM tends to lose its heterogeneity, becoming a simpler, smoother and more homogeneous structure. These results are consistent with quantitative signal analysis[3].
NM volume: The decrease in volume reflects possible neuronal degeneration or loss of NM pigment. In addition, a positive correlation was observed between NM volume and striatal BP on Dat Scan images. These parameters could therefore represent a relevant proximal Dat Scan marker for AMS, reflecting underlying dopaminergic denervation. Our study reveals significant differences in R2* values, NM volume and striatal BP between our groups. These results suggest that the morphological approach, combined with the analysis of magnetic relaxation and hypofixation properties, is promising for the identification of specific biomarkers of AMS. In addition, the radiomic features extracted from the radiomic analysis offer innovative prospects for early detection and characterisation of the disease.
Camille BRUN (Toulouse), Germain ARRIBARAT, Patrice PERAN, Sabrina HOUIDEF
13:30 - 15:00
#47865 - PG542 Structural alterations of Human hypothalamic nuclei in isolated REM sleep behavior disorder .
PG542 Structural alterations of Human hypothalamic nuclei in isolated REM sleep behavior disorder .
Isolated REM sleep behavior disorder (iRBD) is considered a potent early marker of synucleinopathies, such as Parkinson’s disease (PD). Studies mapping brain circuits in rodents have shown that the hypothalamus (HT) plays a central role in regulating sleep and wakefulness, containing both sleep-promoting and wake-promoting neurons that act as 'switches' for sleep-wake transitions [1]. However, few studies account for the HT when investigating sleep-related disorders in humans. In this preliminary study, we combined the use of high-resolution quantitative 3D-T1 MRI sequences with a super-resolute atlas of HT nuclei [3] to uncover macro and microstructure variations of HT nuclei, and their association with the disease duration in well-characterized cohorts of iRBD participants.
We included 8 controls (age: 67 ± 9,22 [46-76]) and 19 iRBD participants (age: 72 ± 7,45 [50-77]; duration: 60 months ± 45,79 [15-156]) meeting the diagnostic criteria for iRBD [4] with polysomnographic confirmation. Subjects were scanned with a 7T MR scanner (TERRA, Siemens) using a (1TX/32RX) Head coil (NOVA). Anatomical data was obtained using a 3D-MP2RAGE sequence (TR=5.000ms/TE=3ms/TI1=900ms/TI2=2.750ms, 256 slices, 0.6mm isotropic resolution, TA= 10min12s). Unbiased T1 maps and T1-w-UNIDEN images were generated after B1+ inhomogeneity correction [5]. T1-weighted volumes; as well as the Neudorfer template/atlas (0.5mm isotropic voxel) [3], were cropped in a similar manner, before an optimal spatial registration using the antsRegistration procedure [6] was performed (Figure 1) [7]. Volumes and T1 values of HT nuclei were extracted using ITKSNAP [8]. The volumes of HT nuclei were normalized by intracranial volume (ICV) [9] prior to group comparisons. The Wilcoxon test (JMP®, Version 18. SAS Institute Inc., Cary, NC, 1989–2023.) was performed to evaluate the modulation of macro and microstructure of individual HT nuclei by iRBD. Partial correlations were performed to study the associations between HT nucleus metrics and disease duration. No significant differences in age (puncorr= 0.096), ICV (puncorr= 0.097) or whole HT volume (zscore: 0,472; puncorr= 0,318) were observed between the two groups. Relative to controls, iRBD participants showed lower T1 values in the left dorsal periventricular hypothalamus (DPEH) (zscore: -1.861; puncorr= 0.031) and right zona incerta (ZI) (zscore: -1.75; puncorr= 0.04) and lower volume of right dorsomedial hypothalamic nucleus (DM) (zscore: -1.694; puncorr= 0.045) (Figure 2). However, iRBD participants showed higher volume of the left paraventricular nucleus (Pa) (zscore: 2.31; puncorr= 0.011) and right medial preoptic nucleus of the hypothalamus (MPO) (zscore: 2.194; puncorr= 0.014) (Figure 2). Partial correlation accounting for age showed significant association between the disorder’s duration and the T1 value of the left DPEH (β: -0.392, puncorr= 0.047) and the volume of the right DM (β: 0.414, puncorr= 0.036). In this preliminary study (control recruitment still ongoing), we identified alterations in the microstructure and macrostructure of several HT nuclei involved in sleep homeostasis.
First, T1 values of the right ZI and left DPEH were found to be lower in iRBD participants. The decrease in T1 values supports the hypothesis of iron accumulation associated to neurodegeneration of the nuclei. Interestingly, ZI promotes sleep through a subset of GABAergic neurons that are active during REM sleep, anticipate sleep onset, and can shift states of consciousness when stimulated [10-11]. DPEH, for its part, plays a central role in autonomic regulation due to its extensive connections throughout the HT and brainstem. This might contribute to neurogenic orthostatic hypotension frequently observed in synucleinopathies.
Macroscopically, we observed a significant atrophy of the right DM, a HT nucleus contributing to the proper regulation of the circadian sleep cycle [14]. In contrast, we observed increases in the volume of the right MPO and the left Pa. The MPO plays a crucial role in sleep regulation by housing GABAergic sleep-active neurons that inhibit arousal-promoting regions like the locus coeruleus, thereby promoting the onset and maintenance of both REM and NREM sleep [12]. The Pa participates in the promotion and maintenance of wakefulness [13]. Finally, we found a significant correlation between the disease duration and the T1 values of the left DPEH and the volume of the right DM, supporting their impairment in the neurodegenerative process. Our findings suggest that structural changes in hypothalamic sleep-regulating regions may contribute to iRBD and serve as early biomarkers of its underlying pathology. By extending insights from rodent studies to humans, our research highlights the hypothalamus' role in sleep homeostasis and underscores the need for early biomarkers, as most iRBD patients develop synucleinopathies within ten years [15].
Coleen ROGER (Marseille), Camille COMET, Hugo DARY, Marie-Pierre RANJEVA, Maxime GUYE, Jean-Philippe RANJEVA, Alexandre EUSEBIO, Stephan GRIMALDI
13:30 - 15:00
#47320 - PG543 Structural and microstructural characterization of corpus callosum integrity in paediatric-onset Huntington Disease.
PG543 Structural and microstructural characterization of corpus callosum integrity in paediatric-onset Huntington Disease.
Huntington disease (HD) is an autosomal dominant disorder, caused by expanded Cytosine–Adenine–Guanine (CAG) repeated mutations (>35 CAGs) in the huntingtin gene (HTT)[1,2], diagnosed by genetic testing. The typical onset occurs in adulthood (adult-onset HD, AOHD) while pediatric-onset HD (POHD)[3,4,5] emerges before age 18, when CAG repeats exceed 55-60, with symptoms such as rigidity and bradykinesia, contrasting with chorea, observed in AOHD. Research suggests more severe subcortical damage in POHD, while cortical involvement occurs in later stages of the disease; extensive evidence also suggests severe white matter (WM) involvement in HD, whether this happens to a different extent between AOHD and POHD remains largely unknown. This study focuses on comparing the structural integrity of the corpus callosum (CC) in POHD, AOHD, and healthy controls (HC) to explore potential differences in cortical region impact between adult and pediatric HD.
We enrolled 21 patients with HD (5 POHD and 16 AOHD) and 27 HC stratified by age to serve as control groups for AOHD and POHD respectively (AHC, 16F/2M; mean age ± standard deviation = 51.9 ± 12.2 years; PHC, 7F/2M; mean age ± standard deviation = 25.3 ± 2.1 years).
Each participant underwent 3T PET/MRI (Biograph mMR, Siemens Healthineers, Forchheim, Germany) using a 16-channel PET-transparent head/neck coil and the protocol included: whole-brain T1-weighted and echo-planar diffusion-weighted imaging sequences.
All images were processed using Freesurfer, FSL and an in-house software to analyze the corpus callosum (CC) profile[6,7]. Using Freesurfer’s standard recon-all pipeline, we extracted for each subject a binary mask of the CC, as well as the volumes from the 5 subdivisions equally spaced along the postero-anterior direction (Figure 1A) and these subdivisions roughly resemble Witelson’s parcellation of the CC as well as the histological distribution of callosal fiber types (Figure 1B and C). All CC masks were also visually checked for segmentation errors that, if present, were manually corrected using Fsleyes. The final mask was then fed to the in-house software for automated extraction of the callosal thickness profile, already described and validated in Caligiuri et al. previous[6,7].
Diffusion-weighted scans were processed using FSL’s DTIFit to obtain maps of fractional anisotropy (FA) and mean diffusivity (MD). For each subject, FA map was nonlinearly registered in subject’s T1-space using FLIRT and FNIRT tools of FSL; the estimated transformation matrix was then saved and applied to MD map, thus obtaining anatomical correspondence across different images of the same subject. In all analyses we investigated the volumes from the 5 CC subdivisions, obtained via Freesurfer, as well as thickness, FA and MD values from all 50 midsagittal CC points.
Statistical analysis was conducted on the structural and diffusion metrics across subdivisions, with comparisons between i)AHC and AOHD, ii)PHC and POHD, and iii)AOHD and POHD using ANOVA or non-parametric tests based on residual normality (significant threshold was set to p=0.05). Volume reductions in AOHD compared to AHC were observed in posterior(p=0.004), middle posterior(p=0.007), central(p=0.004) and middle anterior(p=0.05) CC (Figure 2) while when comparing the two HD forms, AOHD showed decreased volume in posterior CC (p=0.009) compared to POHD (Figure 2). No significant differences were found in thickness.
When comparing FA values, we observed decreases of the metric along the entire profile between AOHD and AHC; no differences were observed between POHD and PHC; FA decreased in middle posterior(p=0.05), central(p=0.01) and anterior(p=0.05) CC (Figure 3) between AOHD and POHD.
When comparing MD values, we observed a large increase in posterior(p<0.001), middle posterior(p<0.001), central(p<0.001) and middle anterior(p=0.002) CC between AOHD and AHC; no significant differences between POHD and PHC nor in AOHD versus POHD (Figure 4). To the best of our knowledge, this is the first report of DTI measures in POHD, hence these findings provide precious insights regarding in vivo disease-related alterations across cerebral WM. Overall, our results suggest that CC is substantially preserved in POHD compared to both PHC and AOHD patients with similar disease severity. On the other hand, all structural and diffusion measures were altered in AOHD across most of the antero-posterior profile of CC compared to age- and sex-matched HC (AHC). Our data suggest the CC to be a key neuropathological structure in the development of HD and could help to differentiate AOHD from POHD due to different characterization along CC profile. These could stimulate further research investigating the CC as a potential brain biomarker of HD disease.
Maria Celeste BONACCI (Catanzaro, Italy), Maria Eugenia CALIGIURI, Ferdinando SQUITIERI, Umberto SABATINI
13:30 - 15:00
#47577 - PG544 A Machine Learning Approach Based on Quantitative MRI White Matter Lesion’s Features for the Identification of Progression Independent of Relapse Activity in Multiple Sclerosis.
PG544 A Machine Learning Approach Based on Quantitative MRI White Matter Lesion’s Features for the Identification of Progression Independent of Relapse Activity in Multiple Sclerosis.
Quantitative MRI (qMRI) techniques are sensitive to microstructural tissue damage and can provide insights into disability accumulation in neurodegenerative and demyelinating conditions such as Multiple Sclerosis (MS). Progression independent of relapse activity (PIRA) is a slow accumulation of disability occurring in the absence of inflammatory acute attacks, and represents the major cause of physical and cognitive disability in MS. We hypothesize that qMRI enables the detection of the subtle neurodegenerative/neuroinflammatory changes white matter lesions (WML) associated with PIRA in MS. Thus, in this study, we assessed whether a machine learning (ML) based approach on qMRI-derived WML features can distinguish MS patients with and without PIRA.
MS patients with (PIRA) and without PIRA (No-PIRA) were neurologically identified based on longitudinal Expanded Disability Status Scale (EDSS) follow-up. Mann-Whitney U-test was used to assess demographic differences across the two patients’ classes. All patients were acquired on a 3T Philips (Elition S) with a multiparametric protocol to estimate semi-quantitative and quantitative MRI features. The MR acquisition protocol and qMRI metrics estimation approaches are shown in Table 1. WML were manually segmented on Fluid Attenuated Inverse Recovery images (3D FLAIR: TR/TE: 3000/260 ms, voxel size 1 mm³ iso) and divided in three lesion types based on their location: isolated, juxtacortical and confluent lesions (Fig.1). Quantitative features were extracted from each lesion and summarized using median values. QSM images were used to identify Paramagnetic Rim Positive Lesions (PRL+), lesions identified by hyperintense iron accumulation at the lesion border and included as a categorical feature in the model. After a model selection strategy implemented in Pycaret [9], the Extra Trees Classifier model was identified as the one maximizing the accuracy metric. A robust evaluation was conducted through 1000 random permutations used for the initial subject-wise split into test and train datasets, with fixed subject percentages of 13% and 87%, respectively. In each permutation the WML of 12 random subjects were reserved for testing, while the WML of the remaining training subjects were pooled balancing the distribution of both lesion classes (PIRA=1; NoPIRA=0) and lesion types using a stratified undersampling. Extra Trees classifier was applied to predict lesion classes in each permuted test set. Performance was evaluated at the lesion level and at the subject level using major voting, where the predicted class for each subject was determined by the most frequent prediction among their lesions. Feature importances were extracted from the model in each permutation. Final performance metrics (lesion and subject levels) and feature importances were then calculated by averaging results across all 1000 permutations to provide a robust aggregate assessment. A total of 1,634 WMLs were segmented from 96 MS patients. Table 2 outlines detailed dataset description. PIRA patients were older, had longer disease duration, and greater disability (EDSS) than NoPIRA patients (p<0.001 for all comparisons). The obtained model achieved: a lesion-level accuracy of 69.6% averaged across the permutations (mean of 205 lesions predicted for each permutation) and a subject-level accuracy (major voting) of 69.7% on average (12 subjects evaluated per permutation). Figure 2 shows the top 10 most important qMRI features averaged from the Extra Trees classifier across all 1000 permutations. Utilizing a machine learning approach with an Extra Tree classifier, qMRI lesion features demonstrated the ability to effectively discriminate WML associated with PIRA in Multiple Sclerosis. This framework, by aggregating lesion-level predictions through a majority voting strategy, also enabled classification at the subject level. Despite including only white matter qMRI features and addressing the particularly challenging task of detecting subtle disability progression, our approach achieved a good performance. These results highlight the potential of qMRI-derived metrics to capture tissue microstructural differences which may contribute to enhance the understanding of mechanisms most representative of progression in MS. qMRI features enabled effective ML classification of PIRA patients . These findings suggest that incorporating advanced imaging biomarkers into routine clinical workflows could support more precise monitoring of disease activity. Future work should validate the model across independent cohorts, explore the longitudinal evolution of these qMRI features and understand the neuropathological correlates of these alterations to develop in-vivo markers specific to progression.
Francesco GUARNACCIA (Verona, Italy), Agnese TAMANTI, Nicola DALL'OSTO, Valentina CAMERA, Laura PASTORE, Rachele BONETTI, Samuele QUAGLIOTTI, Teresa MALTEMPO, Arianna CAVAGNA, Sophia CAMERER, Marco CASTELLARO, Roberta MAGLIOZZI, Francesca Benedetta PIZZINI, Massimiliano CALABRESE
13:30 - 15:00
#47728 - PG545 Glymphatic system impairment in relapsing-remitting multiple sclerosis: Diffusion along perivascular spaces index correlation with volume fraction metrics using multishell diffusion MRI.
PG545 Glymphatic system impairment in relapsing-remitting multiple sclerosis: Diffusion along perivascular spaces index correlation with volume fraction metrics using multishell diffusion MRI.
The glymphatic system (GS), which facilitates the cerebral waste clearance[1], can be evaluated noninvasively using diffusion tensor imaging (DTI) along perivascular spaces (ALPS) index.[2] Previous studies showed an reduced ALPS index in patients with relapsing remitting multiple sclerosis (RRMS) compared to healthy controls[3]. Also, microstructural changes in normal appearing white matter have previously been demonstrated in RRMS patients.[4] However, the impact of the changes in microstructure on the ALPS index have not yet been investigated in RRMS patients.
Participants and MRI-Application
In 42 RRMS cases (35.2 ± 8.5 years; 11 males, no relapse for 3 months, expanded disability status scale (EDSS) < 4). We performed a multishell-DTI with 82 diffusion directions and 5 b-values (0, 300, 700, 1000, 2000 s/mm2) using a 64-channel head coil in 3T (Magnetom Vida, Siemens Healthiness Erlangen, Germany). The local ethical commission approved the study.
Post processing
The DWI data underwent 5 corrections steps in the post-processing stage: truncation artefact by mrtrix3 [5, 6], motion artefacts, magnetic field inhomogeneities, eddy currents and variation in spatial intensity (FSL group: FAST, topup, Eddys). [7, 8, 9]
Afterwards, we did a multi-shell multi-tissue constrained spherical deconvolution (MSMCSD) analysis to estimate fiber orientation distribution[10]. Whole-brain tractography was then generated using 2nd order integration over fiber orientation distributions algorithm implemented in Mrtrix3.[11]. Then, we used a stick-zeppelin-ball model [12] to calculate the intracellular (IC), extracellular (EC) and isotropic (ISO) volume fraction (VF) using COMMIT.[13]
ALPS index calculation and correlation with VF
For analysis, we loaded the following images in DSI studio: (1) susceptibility weighted image (SWI) to identify that the vessels are perpendicular to the ventricles. (2) FLAIR to ensure, that the regions of interest (ROI) were not placed in white matter lesions, and (3) the different volume fraction images for ICVF, ECVF, and ISOVF.
We placed the ROIs for the ALPS index calculation in the projection (corona radiata) and association areas (superior longitudinal fasciculus), avoiding white matter lesions (Fig. 1).
We calculated the left and right ALPS index using the diagonal elements of tensor matrix in DSI studio[14] .
We used jamovi for statistical analysis. Spearman’s correlations between ICVF, ECVF, and ISOVF of association and projection fibers and the ALPS index were examined in both hemispheres. The ALPS index was 1.47 ± 0.18 in the left and 1.42 ± 0.18 in the right brain hemisphere, showing no significant interhemispheric differences.
A significant moderate positive correlation was found between in ICVF and the ALPS index in projection (Spearman's ρ = 0.5, p < 0.001) and association area (Spearman's ρ = 0.5, p < 0.001) of the left hemisphere. For the association fiber of the right hemisphere, ICVF and ALPS index showed a significant weak to moderate correlation (Spearman's ρ = 0.35, p = 0.02).
There is no significant correlation between right ALPS index and ICVF in the projection fibers (Spearman’s ρ = 0.28, p = 0.08).
For the ISOVF and the ALPS index, there was a moderate correlation in the left projection fibers (Spearman’s ρ = -0.44, p = 0.004). No further significant results were found.
The significant results of the correlation between the ALPS index and the ICVF are shown in Fig. 2. The ALPS index did not correlate with either ECVF or ISOVF, suggesting no evidence for accumulation of osmotically active waste products.
Given the positive correlation between ICVF and the ALPS index (Fig. 2), we suggest that a reduced ICVF could be indicative of microstructure damage[4]. This may be associated with a lower ALPS index, reflecting impaired GS function in RRMS[2].
Our patient cohort consisted of patients with a low EDSS. This could explain the lack of further associations. Also, we suggest that a larger sample size to provide more definitive conclusion. We demonstrate a relationship between changes in the IC compartment and the ALPS index, which may serve as an indication of GS impairment in RRMS. This can help to understand the influence of microstructure changes in the meaning of the ALPS index.
Janina KREMER (Lübeck, Germany), Andreas Martin STROTH, Katja HUMMEL, Norbert BRÜGGEMANN, Philipp J. KOCH, Peter SCHRAMM, Patricia ULLOA
13:30 - 15:00
#48022 - PG546 Application of BBB-ASL in MS: Initial Experience.
PG546 Application of BBB-ASL in MS: Initial Experience.
Multiple Sclerosis is an autoimmune disease of the central nervous system and is characterised by demyelination and neurodegeneration [1]. Breakdown of the blood-brain barrier (BBB) is one of the hallmarks of MS [1]. T1-weighted (T1w) MRI acquired after an injection of gadolinium-based contrast agents (GBCA) has been utilized to identify leakage of the BBB in MS [2]. However, due to gadolinium’s higher molecular weight, post-contrast T1w MRI might miss subtle BBB changes. BBB Arterial Spin Labelling (BBB-ASL) MRI is a new technique to assess BBB water permeability, and it has been studied in healthy volunteers and patients with brain tumors [3-5]. This study is an initial effort to investigate BBB permeability in MS patients, focusing on both lesions and normal-appearing white matter (NAWM) using BBB-ASL.
Seven people with MS (pwMS) and two healthy volunteers were scanned on a clinical 3T MRI scanner (Prisma, Siemens Healthineers, Erlangen, Germany) using a 32-channel head coil. A combination of single-TE and multi-TE Hadamard-encoded pseudo-continuous (pCASL) sequences, implemented using the vendor-independent MRI framework gammaSTAR [7], with 3D GRASE readout and two FOCI inversion pulses to suppress background with T1 values of 700 and 1400 ms, was used. A single-TE Hadamard-8 matrix was acquired with a sub-bolus duration of 400 ms, post-labeling delays (PLD [ms]) of 600 and 800, TE=13.2 ms, TR=4000 ms, resulting in two sets of seven inflow times (TI [ms]) [1000:400:3400] and [1200:400:3600], respectively. Additionally, a multi-TE Hadamard-4 matrix was acquired with a sub-bolus duration of 1000 ms, PLD of 500 ms, TR 4500 ms, eight TEs [13.8:27.6:207 ms], resulting in datasets with three TIs [1500:1000:3500 ms]. Pre- and post-contrast 3D T1w MPRAGE (TR=2300 ms, TE=2.26 ms, TI= 900ms, flip angle=8, slice thickness=1 mm) were acquired as structural references. A 3D fluid attenuated inversion recovery (FLAIR) sequence (TR=5000 ms, TE=388 ms, slice thickness=0.9 mm) was acquired. Cerebral blood flow (CBF) and water exchange time (Tex) maps were quantified using ExploreASL [8]. Bias field correction and brain extraction were performed on FLAIR images using FSL FLIRT and BET [9-10]. The Lesion Prediction Algorithm (LPA) of the Lesion Segmentation Toolbox (LST) was used to obtain lesion masks automatically [11]. White matter (WM) and gray matter (GM) tissues were segmented on T1w images using CAT12. The lesion masks of the patients were excluded from the corresponding WM masks to obtain normal-appearing WM (NAWM) masks of the patients. All masks were registered to the ASL space using SPM12. The histogram values of the CBF and Tex maps were assessed using MATLAB 2024 (Natick, MA). A Wilcoxon signed-rank test was used to compare CBF and Tex values between the histogram values of the FLAIR lesions and NAWM, using a single representative value per subject per region. This study included seven pwMS, a mean age of 21.29±11.01y, F/M=5/2 (seven relapsing remitting MS (RRMS)), and two healthy volunteers with a mean age of 34±8.5y, two females. Table 1 summarizes the mean, 10th percentile, and 90th percentile values of the CBF and Tex maps in volunteers. The mean CBF values of volunteers in NAWM were 25.7 and 21.05 ml/100g/min, while the mean Tex values in NAWM were 154.2 and 159.6 ms. Figure 1 shows the mean, 10th, and 90th percentiles of the CBF values at the lesion and NAWM. The mean CBF values in the lesions (31-74 ml/100g/min) were higher than those in the NAWM (26-68 ml/100g/min)(p=0.0156). Similarly, the 10th percentile values at the lesion (9-35 ml/100g/min) were elevated compared to NAWM (7-21 ml/100g/min)(p=0.0312). The 90th percentile of CBF values was slightly higher in the lesions (52-142 ml/100g/min) than in the NAWM (54-122 ml/100g/min)(p=0.0156). Figure 2 presents the mean, 10th, and 90th percentiles of the Tex values in lesions and the NAWM. Overall, the Tex values of NAWM were lower than those of lesions. While the difference in Tex values did not reach statistical significance, the trend between the values was observed. In this initial study, the BBB-ASL method was used to evaluate CBF and Tex values in the lesions of seven pwMS, providing a non-invasive approach for assessing BBB permeability. The finding of increased CBF in FLAIR lesions compared to NAWM is consistent with previous literature [12]. Although the increased Tex values in FLAIR lesions compared to NAWM align with prolonged mean transit time (MTT) findings reported in the literature [13], this difference did not reach statistical significance and needs further investigation. Future studies will aim to evaluate the utility of the BBB-ASL method for assessing perfusion metrics in a larger patient cohort, with consideration of lesion activity and T2 heterogeneity. The BBB-ASL method can non-invasively capture the difference between lesional and normal-appearing WM in MS patients.
Ayse Irem CETIN (Istanbul, Turkey), Gulce TURHAN, David R VAN NEDERPELT, Ahmed Serkan EMEKLI, Amnah MAHROO, Beatriz E. PADRELA, Simon KONSTANDIN, Daniel Christopher HONKISS, Nora-Josefin BREUTIGAM, Vera KEIL, Frederik BARKHOF, Klaus EICKEL, Henk MUTSAERTS, Matthias GÜNTHER, Dilaver KAYA, Alp DINÇER, Jan PETR, Esin OZTURK-ISIK
13:30 - 15:00
#46770 - PG547 Integrating BMAT and FA Imaging for Improved Classification of Multiple Sclerosis: A MachineLearning Perspective.
PG547 Integrating BMAT and FA Imaging for Improved Classification of Multiple Sclerosis: A MachineLearning Perspective.
Multiple sclerosis (MS) affects both cognitive and physical functions, making an accurate diagnosis essential for effective management (1). While cognitive tests like the Brief Memory and Attention Test (BMAT) help track delayed working memory (2), they may miss early cognitive changes and are influenced by examiner expertise. Magnetic resonance imaging (MRI) is the most effective tool for diagnosing MS, but does not always correlate with clinical symptoms (3). To address these limitations, this study combines cognitive assessment (BMAT), MRI-derived Fractional Anisotropy (FA), and machine learning (ML) techniques (4–6) to enhance MS classification by cognitive status and develop a more robust diagnostic framework.
Diffusion-weighted and T1-weighted images were acquired in a 3T MRI scanner (Philips Ingenia) in 41 HC (59% female) and 58 RRMS patients (67% female). The local ethics committee approved the study, and RRMS patients were diagnosed according to the 2017 McDonald's criteria. All patients were evaluated with an Expanded Disability Status Scale (EDSS) and verbal memory assessment using BMAT. All test scores were normalized in Z-scores. Using the Z-score, patients and healthy controls were categorized as HC-CP and RRMS-CP with a Z-score ≥ -1.5 and RRMS-CI with a Z-score < -1.5. Diffusion-weighted images were processed to obtain FA maps using DTI. T1-weighted images were used as an anatomical reference. All preprocessing steps were performed in SPM12. We used the LNAO-SWM79 U-fiber atlas as a mask to obtain the mean FA map for each subject's U-fiber. The preprocessing step was done through an in-house MATLAB toolbox.
An ML model was designed to predict those regions whose FA values adequately discriminate between HC-CP, RRMS-CP, and RRMS-CI classes. The model was built using Python and employed Random Forest (RF) with Sequential Forward Selection (SFS) for feature selection. The RF’s hyperparameters and evaluation of the performance of the classification model were implemented using stratified 5-fold cross-validation. The RF fine-tuned parameters that minimized the mean absolute error were: max_depth = 10, max_leaf_nodes = 5, min_samples_split = 11, and n_estimators = 299. We evaluated feature selection and the RF performance according to the number of features selected and their importance with the higher score with optimal features. Figure 2 shows the accuracy performance of the RF model concerning the most relevant hemodynamic features selected by SFS. U-fibers that connect different brain regions categorized forty-four (Figure 3): frontal lobe regions connected with (rostral anterior cingulate, medial orbitofrontal, superior frontal area, pars triangularis and pars opercularis), parietal lobe regions with (inferior parietal, superior parietal, and supramarginal gyrus), temporal lobe regions with (middle temporal, superior temporal, and inferior temporal), insular and cingulate with (insula and posterior cingulate), and occipital lobe with fusiform area. They resulted in an accuracy of 80.89 ± 7.88% (precision: 91.01 ± 2.14%, recall: 80.98 ± 5.01%, F1-score: 85.05 ± 5.01%). The RF decision trees for each classification problem are shown in Figure 4. The identified features from key brain regions are critical in improving MS classification accuracy. By leveraging the diverse functions associated with areas such as the frontal lobe (involved in executive functions and emotional regulation), parietal lobe (related to sensory integration and spatial awareness), and temporal lobe (crucial for memory and language processing), we gain deeper insights into the cognitive and neuroanatomical impacts of MS.
The involvement of insular and cingulate regions highlights the importance of emotional and interoceptive awareness in MS patients, as these areas are linked to the disease's psychological and cognitive challenges (7, 8). Additionally, the role of the fusiform area underscores the significance of visual processing, which may be affected in MS (7).
These findings align with previous evidence suggesting that MS-related cognitive deficits are associated with widespread structural and functional brain alterations, particularly in regions supporting high-order cognitive and emotional functions. The classification of MS based on neural features offers valuable insights into disease progression and underlying neurobiological mechanisms. This approach can enhance early diagnosis and guide treatment strategies. Integrating machine learning techniques with neuroimaging data holds strong potential for developing targeted interventions aimed at mitigating cognitive decline in MS patients.
Cristian MONTALBA, Cristian MONTALBA (Santiago, Chile), Pamela FRANCO, Raúl CAULIER-CISTERNA, Macarena VASQUEZ, Claudia CÁRCAMO, Ethel CIAMPI, Marcelo ANDIA
13:30 - 15:00
#47807 - PG548 Assessment of microcirculatory changes in normal-appearing white matter and in the gray matter of the brain in patients with multiple sclerosis by perfusion MRI.
PG548 Assessment of microcirculatory changes in normal-appearing white matter and in the gray matter of the brain in patients with multiple sclerosis by perfusion MRI.
Purpose: to evaluate perfusion changes in normal-appearing white matter (NAWM) and in the gray matter of the brain with demyelinating lesions of the central nervous system using the method of dynamic susceptibility contrast (DSC).
The MR study was carried out on a MR-scanner "Ingenia" ("Philips") 3 Tesla using the method of dynamic susceptibility contrast (DSC). The study included 30 healthy volunteers and 80 patients with demyelinating disease of the central nervous system (9 patients with CIS, 66 patients with RRMS and 5 patients with SPMS) over the age of 18 up to 48 years (average age was 34.6 ± 8.02 years). Quantitative and qualitative assessment of CBF, CBV, MTT, TTP in normal-appearing white and gray matter in the frontal, parietal, temporal and occipital lobes of the brain. In all groups in NAWM, a significant decrease in CBF and CBV was observed in all lobes of the brain, and the severity of these changes increases with the progression of the disease: in patients with CIS CBF is reduced to 13.7% and CBV to 7.3%; the most pronounced decrease in perfusion was observed in patients with SPMS: CBF by 40% and CBV by 24.8% (p <0.001); with a moderate increase in TTR and MTT by 15%.
Similar changes are visualized in the gray matter of the brain: in patients with CIS CBF is reduced to 14.7% and CBV to 6.9%; the most pronounced decrease in perfusion was observed in patients with SPMS: CBF by 36.5% and CBV by 28.2% (p <0.001); with a moderate increase in TTR and MTT by 15.8%. In patients with CIS, the increase in perfusion parameters is more pronounced, which indicates the predominance of inflammatory changes, which likely cause clinical manifestations. At the same time, in patients with a secondary progressive course, in whom an increase in the degree of disability is clinically observed, hypoperfusion is observed, probably associated with hypoxia developed against the background of prolonged inflammation and with a reduced metabolic demand, which indicates neurodegeneration.
Thus, we can conclude that in the initial stages of the disease, inflammatory changes predominate, while as the disease progresses, neurodegeneration predominates. Assessment of cerebral perfusion allows you to take a fresh look at the role of the vascular component in the pathogenesis of multiple sclerosis. Perfusion data complements routine MRI and provides a comprehensive assessment of changes in brain matter.
We thank the Russian Science Foundation for supporting this work (№ 23-15-00377).
Liubov VASILKIV, Yulia STANKEVICH, Olga BOGOMYAKOVA, Denis KOROBKO, Vladimir POPOV, Nadezhda MALKOVA, Andrey TULUPOV (Novosibirsk, Russia)
13:30 - 15:00
#47687 - PG549 Non-contrast quantitative perfusion MRI in multiple sclerosis.
PG549 Non-contrast quantitative perfusion MRI in multiple sclerosis.
Non-contrast MR perfusion (arterial spin labeling, ASL) can detect areas of altered cerebral perfusion in patients with multiple sclerosis (MS), even in the absence of focal lesions [1]. This method offers advantages such as non-invasiveness [2] and short acquisition time. The use of ASL in MS patients is important for diagnosis, treatment strategy, and disease prognosis, but this area remains insufficiently studied [3]. The purpose is to optimize the algorithm and investigate cerebral perfusion changes using ASL in patients with multiple sclerosis compared to a control group.
This prospective study included 15 patients with MS and 15 age- and sex-matched healthy controls. All participants underwent 3.0T MRI (Philips Ingenia) with a standard protocol (T1-WI, T2-WI, FLAIR, DIR, post-contrast T1-WI) supplemented by pseudocontinuous arterial spin labeling (pCASL, FOV:240x240x99; TR:4550; TE:16; LD:1800; PLD:1800) for cerebral perfusion assessment in ml/100g/min. To address computational challenges in ASL quantification, we developed an optimized processing pipeline incorporating Radiant, FSL (BASIL) and MriCroGL. We calculated cerebral perfusion in the BASIL (FSL) program. Nonparametric statistical methods were used for data analysis. Healthy controls demonstrated median gray matter perfusion of 51.9 mL/100g/min (IQR: 51.4-53.6) and white matter perfusion of 16.8 mL/100g/min (Fig. 1, IQR: 15.1-18.8). In MS patients, gray matter perfusion was significantly reduced (43.7 mL/100g/min, IQR: 42.8-44.5), as was white matter perfusion of 14.7 mL/100g/min (Fig. 2, IQR: 14.0-15.2) both p<0.001 vs. controls (Fig. 3, 4). Focal demyelinating lesions showed severe hypoperfusion (9.7 mL/100g/min, IQR: 5.3-13.2). Our study demonstrates that optimized pCASL reliably quantifies both focal and diffuse perfusion abnormalities in MS, revealing significant hypoperfusion in gray matter (15.8%) and normal-appearing white matter (12.5%) compared to controls. These findings align with growing evidence of microvascular dysfunction as a key pathophysiological mechanism in MS [4, 5]. The observed perfusion reductions in structurally intact tissue may precede visible lesions, suggesting ASL’s potential as an early biomarker for disease progression. Future studies should correlate ASL metrics with clinical disability scores and explore longitudinal perfusion changes. The developed pCASL processing algorithm enables comprehensive perfusion assessment in multiple sclerosis patients, quantifying both demyelinating lesions and normal-appearing white matter. Our results demonstrate a significant (p<0.001) reduction in perfusion within normal-appearing white matter (12.5%) and gray matter (15.8%) compared to healthy controls.
We thank the Russian Science Foundation for supporting this work (№ 23-15-00377).
Andrey TULUPOV (Novosibirsk, Russia), Vladimir POPOV, Lyubov VASILKIV
13:30 - 15:00
#46082 - PG550 MRI-based mapping of disease evolution in a preclinical model of multiple sclerosis.
PG550 MRI-based mapping of disease evolution in a preclinical model of multiple sclerosis.
Experimental autoimmune encephalomyelitis (EAE) is well-established model characterized by presenting neurological deficits that closely resemble those observed in individuals with multiple sclerosis. The aim of this study was to characterize the temporal evolution of brain and spinal cord pathology in EAE using ex-vivo magnetic resonance imaging (MRI).
EAE was induced using the 35-55 myelin oligodendrocyte glycoprotein peptide. Animals were scored daily for neurological signs on a 6-point scale. At baseline, day 15 (inflammatory phase) and day 50 (neurodegenerative phase), six animals per time-point were euthanized and their brains and the spinal cords were removed and fixed in 4% paraformaldehyde. MRI was acquired on a Brucker 7.0 T system. The protocol included: T1 and T2 multi-echo mapping, diffusion-weighted (fractional anisotropy (FA)), and magnetization transfer ratio (MTR). Quantitative measures were obtained for each modality, based on a parcellated brain atlas and the Spinal Cord Toolbox for spinal grey/white matter. Differences between time points were assessed using ANOVA followed by Tukey’s post-hoc test (p<0.05). All brains except two could be analyzed, while in the spinal cord, measures in partial segments could be obtained in 50% of the animals. The reasons for exclusion were tearing and shearing of the tissue, especially at later time points. In the brain, T1 values significantly increased from baseline to day 15 in 10 out of 14 regions (5-8%, p<0.01 in all regions), with no further changes by day 50. MTR showed a similar profile, though differences over time were not significant. No changes were measured in T2 mapping. FA showed significant reductions in 5 of 14 regions at day 50 compared to earlier time points (6–13%, p=0.01–0.045). In the spinal cord, FA decreased significantly at day 50 compared to baseline (17% reduction, p=0.003), while no significant differences were found in T1, T2, and MTR. Early T1 elevation in brain regions likely reflects inflammation or edema, while later FA reductions suggest neurodegeneration, which could reflect axonal loss or demyelination. Further studies incorporating histopathological correlation will help to further elucidate and appropiately interpret these findings. Ex-vivo MRI can be processed using established analysis pipelines, offering quantitative insights into EAE progression. This approach is particularly valuable, as it enables histopathological analysis to be performed on the same animal, allowing direct correlation with imaging findings. However, careful tissue handling is essential to preserve data quality.
Laura MARTÍNEZ, Imane BOUTITAH, Arnau HERVERA, Mariam CORIS-ERROUICH, Àlex ROVIRA, Herena EIXARCH, Carmen ESPEJO, Deborah PARETO (Barcelona, Spain)
13:30 - 15:00
#47611 - PG551 Deep Learning for Multiple Sclerosis Prognosis: Longitudinal MRI and Multimodal Data Integration to Predict Disability Progression.
PG551 Deep Learning for Multiple Sclerosis Prognosis: Longitudinal MRI and Multimodal Data Integration to Predict Disability Progression.
Multiple Sclerosis (MS) is a chronic, neuroinflammatory disease of the central nervous system and a leading cause of disability in young adults. It is characterized by heterogeneous clinical trajectories and a variable rate of disability progression. While MRI plays a central role in MS diagnosis and monitoring, predicting disability progression remains a clinical challenge owing to due to the disease's diverse manifestations and multifocal pathology [1]. Advancements in artificial intelligence (AI), particularly deep learning (DL), offers much promise for extracting subtle patterns from multimodal data to improve individualized prognosis. Our project aims to develop a robust, interpretable DL framework capable of predicting MS progression using longitudinal multimodal MRI data and clinical metadata, thereby facilitating personalized treatment strategies.
Our study leverages a rich and thoroughly characterized longitudinal dataset comprising 193 relapsing-remitting MS patients followed yearly over 10+ years for a total of 1536 visits [2]. For each visit, we have access concomitantly to various conventional MRI modalities, such as T1w and T2w, and FLAIR images, alongside clinical data such as the EDSS and MSFC scores, and demographic metadata. All data were acquired at the same center, using harmonized protocols and expert-validated progression labels. The DL model architecture consists of two main components: (1) a spatial feature extraction module using 3D ResNet CNNs to identify representations from MRI data and associate them with clinical scores, (2) a temporal feature extraction module employing bidirectional Gated Recurrent Units (GRUs) with a Time-Aware Attention mechanism to model progression across visits. Training is accordingly performed in two stages: first, learning to associate spatial features with EDSS/MSFC at single time points, followed by prognostic modeling using triplets, with two visits as input, one future visit as target. Interpretability of the model outcomes is ensured via Grad-CAM and saliency maps [3], and generalization is addressed through modular training and balanced sampling. Initial experiments using 865 visits from a subset of 104 patients showed promising results. As shown in Figs. 1 and 2, a model trained on all triplets (N=10166) yielded 86% training and 74% validation accuracy, but suffered from overfitting due to data imbalance. The large variance in the number of total visits between the patients, ranging from 3 to 17 (see Fig. 3) was a cause of concern as it had implications for repetition of data during the training process. After limiting analysis to a maximum of 8 visits per patient (N=4280 triplets), performance improved significantly, achieving 90% training and 88% validation accuracy, with more stable validation loss curves. This highlights the need for balanced data distribution to enhance better generalization of model predictions, especially when dealing with very high dimensional data. The model reliably predicted future confirmed disability accumulation (CDA) using MRI and clinical data from prior visits. Our findings demonstrate the value of integrating longitudinal multimodal MRI and clinical data using deep learning approaches for MS prognosis. The incorporation of MRI patterns alongside clinical scores provides a much-needed holistic representation of the patient’s state, enabling the model to extract meaningful biomarkers more effectively. Modular preprocessing and training pipeline enables reproducibility and scalability to larger datasets. Our next steps will focus on minimizing the issue of overfitting even further and enhancing model generalization, while making optimal use of all the available data. This involves a redesign of the model architecture, to enable the use of modularized components that can be trained individually. Ongoing work explores further developments in uncertainty quantification and explainability to increase clinical trust in our AI-generated predictions. Our study takes a step forward towards precision medicine in MS care. By integrating longitudinal MRI and clinical data into a single predictive framework, it aims to enable an interpretable and scalable solution to predict MS progression at the level of individual patients. By employing a variety of clinical data alongside EDSS, such as the MSFC and Rao’s Brief Repeatable Battery [4], completed by MRI-based measures for the patients on each visit, our models would be better trained to capture various forms of disability accumulation. The objective is to target deployment as a clinical decision support tool, with interactive visualizations allowing neurologists to explore lesion-level contributions to predicted outcomes. This work has strong implications for early intervention, treatment planning, and patient counseling in MS.
Shailesh APPUKUTTAN (Marseille), Adrien AMBERTO, Bertrand AUDOIN, Muhammad BILAL, Mounir Mohamed EL MENDILI, Ismail ZITOUNI, Audrey RICO, Hugo DARY, Maxime GUYE, Jean-Philippe RANJEVA, Jean PELLETIER, Wafaa ZAARAOUI, Matthieu GILSON, Adil MAAROUF
13:30 - 15:00
#47725 - PG552 Investigating the link between glymphatic function and white matter microstructure in multiple sclerosis.
PG552 Investigating the link between glymphatic function and white matter microstructure in multiple sclerosis.
The glymphatic system (GS) is a pathway that facilitates the movement of cerebrospinal fluid from the subarachnoid space into brain parenchyma, where it mixes with interstitial fluid to remove metabolic waste through the perivascular space[1]. GS dysfunction contributes to central nervous system pathology and it has been shown that neurological diseases like multiple sclerosis (MS) are linked to an abnormal accumulation of neurotoxic compounds. A non-invasive method to evaluate GS characteristics is the diffusion tensor image analysis along the perivascular space (DTI-ALPS)[2], where the ALPS index is computed from diffusion-weighted images in a region of interest (ROI) in the periventricular white matter (WM), providing a proxy for glymphatic activity and the efficiency of waste clearance from the brain. Despite its utility, traditional DTI-derived metrics have limitations in detecting microstructural changes, especially in regions with complex fiber architecture common to WM connections. Fixel-based analysis (FBA) overcomes these limitations by providing a more detailed estimate of microstructural changes within a population of fibers in a single voxel (fixel), yielding three metrics: fiber density (FD), fiber-bundle cross-section (FC), and their combined effect (FDC). These metrics allow for a more refined assessment of pathophysiological changes, such as fiber loss or atrophy, on a microscopic level, by capturing intra-axonal volume changes (FD) and macroscopic alterations in fiber bundle size (FC). In this study, we conducted and correlated DTI-ALPS and FBA to investigate the association between glymphatic function and WM microstructure integrity in healthy controls (HC), clinically isolated syndrome (CIS) and relapsing-remitting MS (RRMS) patients.
T1- and diffusion weighted image data of 165 adult MS patients (30 with clinically isolated syndrome; 17 of which female; and 135 relapsing-remitting MS, 91 female) and 57 healthy age- and sex-matched subjects (HC) were included in this retrospective study. Scanning was performed on a Magnetom Trio 3 T Siemens scanner. A T1-weighted MPRAGE was used with the following parameters: Voxel size=1x1x1 mm, repetition time (TR)=1900 ms, echo time (TE)=2.52 ms, flip angle=9°, field of view (FOV)=256*256 mm. The T2-weighted echo-planar imaging sequence used to obtain the diffusion-weighted image data had the following parameters: Voxel size 2x2x2.5 mm, TR=9000 ms, TE=102 ms, flip angle=90°. 30 diffusion directions at b=900 s/mm² as well as one image at b=0 s/mm² were acquired. The DTI-ALPS index was calculated automatically for each individual using four spherical ROIs in standard space. FBA metrics (FD, FC, FDC) were computed using the MRtrix3 framework including preprocessing, response function estimation, and population template creation. Group comparisons were performed using connectivity-based fixel-enhancement with non-parametric permutation testing. In addition, correlations between mean FBA metrics and ALPS index values were assessed using Pearson correlation. A multiway ANOVA showed significant differences between the three groups regarding the left (p=0.0047), right (p=0.0151) and mean ALPS index (p=0.0039), and a post hoc test showed, that there was a statistically significant decrease of the ALPS index of RRMS patients compared to healthy controls (Fig. 1) for the left side (p=0.0032), the right side (p=0.0115) and as a mean (p=0.0025). Statistically significant differences between HC and RRMS groups were found for all FBA metrics. Especially regarding FC and FDC, the fixels that differ significantly (after FWE, p<0.05) between the two groups were located in the superior corona radiata (Fig. 2 & 3). Additionaly, left, right and mean ALPS index values showed significant correlations with mean FD, FC and FDC in basal ganglia and thalamus ROIs. Our preliminary results indicate that glymphatic dysfunction, as reflected by a reduced ALPS index, may contribute to or reflect microstructural WM degeneration in MS. In particular, the frequent occurrence of significant fixels in the superior corona radiata, which also plays a role in determining the ALPS index, suggests that there could be a connection between the state of the glymphatic system and the structural integrity of the WM microstructure in said area. The differences between groups found in the basal ganglia and thalamus, which are known to experience atrophic changes over the course of MS [3], may be associated with altered glymphatic function, supporting the hypothesis that axonal damage contributes to MS pathogenesis [4]. However, further work is needed to gain deeper insights into this context. Our findings suggest a link between glymphatic dysfunction and WM degeneration in RRMS. Reduced ALPS index and FBA-derived changes, particularly in the superior corona radiata, basal ganglia, and thalamus, indicate that impaired glymphatic function may contribute to MS-related microstructural damage or vice versa.
Sascha D. SANTANIELLO (Mainz, Germany), Gabriel GONZALEZ-ESCAMILLA, Markus JANKO, Marc A. BROCKMANN, Sergiu GROPPA, Ahmed E. OTHMAN, Andrea KRONFELD
13:30 - 15:00
#47809 - PG553 Evaluation of changes in perfusion and white matter volume in the brain in clinically isolated syndrome.
PG553 Evaluation of changes in perfusion and white matter volume in the brain in clinically isolated syndrome.
Clinically isolated syndrome (CIS) is the initial stage of multiple sclerosis development, has a symptom complex identical to multiple sclerosis, but does not satisfy all the criteria of dissemination in time and space. According to the literature, 30% of patients showed conversion of clinically isolated syndrome to multiple sclerosis within 1 year and about 50% after 5 years; however, Bates et al. found that early treatment of patients with CIS accelerates recovery and can delay the development of multiple sclerosis. Early initiation of effective therapy with DMDs (disease modifying drugs) is a topical issue, which raises the question of when to start it.
Purpose: To assess changes in perfusion and volume of brain tissue in clinically isolated syndrome (CIS).
The MR study was carried out on a MR-scanner "Ingenia" ("Philips") 3 Tesla. The study included 12 healthy volunteers and 6 patients with demyelinating disease of the central nervous system - with clinically isolated syndrome (CIS) and 13 patients with multiple sclerosis. To assess perfusion, the dynamic susceptibility contrast (DSC) method was used. Quantitative and qualitative assessment of CBF and CBV in the white and gray matter of different lobes of the brain. To assess morphometry, the obtained T1-WI and FLAIR images were loaded into an automated system for calculating the volumes of brain structures, based on the segmentation method. The volumes of the white matter of the brain (relWMV) and gray matter (relGMV) were relatively calculated based on the total intracranial volume as a percentage. A moderate correlation was found between the decrease in the volume of white matter of the brain and the severity of focal changes (r-Spearman correlation coefficient 0.4, p≤0.05); compared to CIS, in patients with relapsing-remitting multiple sclerosis in the remission stage and secondary progressive form of MS (SPMS), relCBF was significantly reduced by 32% and 40% (p≤0.05); compared to the CIS group, in patients with RRMS (exacerbation stage) and RRMS (remission stage), the volumes were significantly reduced by 8% and 11% (p≤0.05), respectively. Based on the data obtained, it follows that the pathogenesis of CIS and MS is based on identical processes - changes in perfusion and neurodegeneration, which begin at the earliest stages of the disease.
We thank the Russian Science Foundation for supporting this work (№ 23-15-00377).
Liubov VASILKIV, Yulia STANKEVICH (Novosibirsk, Russia), Olga BOGOMYAKOVA, Denis KOROBKO, Nadezhda MALKOVA, Vladimir POPOV, Andrey TULUPOV
13:30 - 15:00
#46081 - PG554 Assessing the integrity of the optic nerve using conventional magnetic resonance imaging.
PG554 Assessing the integrity of the optic nerve using conventional magnetic resonance imaging.
Optic neuritis is one of the most common manifestations of multiple sclerosis (MS). A complete assessment of the optic nerve requires the acquisition of dedicated sequences. The T1-weighted to T2-weighted ratio (T1/T2) has been proposed as a feasible alternative to assess tissue integrity. Current automated segmentation approaches for T1-weighted magnetic resonance imaging (MRI) do not include the optic nerve parcellation. The aim of this study was to explore the integrity of the optic nerve, based on the T1/T2. For that purpose, a deep-learning approach was developed to segment automatically the optic nerve from conventional 3D T1-weighted MRI.
The cohort included healthy controls (HC, n=18) and people with MS with previous history of optic neuritis (ON, n=16) and without (MS-nONn=17). MRI were acquired in a 3.0 T system (Prisma, Siemens), using a 64-channel head coil. Ground truth masks were generated manually outlining each optic nerve, for all subjects, based on the T1-weighted MRI. For the deep-learning task, a 2D U-Net architecture was implemented (patches 32x32 voxels, four encoding and decoding levels, batch normalization, ReLU activation, and a dropout rate of 0.2). The dataset was divided into training (70%), testing (15%), and validation (15%). To assess the model performance, the generated masks were compared to the ground truths by using the Dice coefficient. In parallel, T1/T2 images were generated and the mean value along the optic nerve profile was obtained over the generated segmentation masks. The mean value of T1/T2 of the optic nerve of the affected eye in ON was compared to the mean value in eyes of HC, MS and optic neuritis fellow eye by using the Kruskal-Wallis test followed by posthoc Dunn’s test (differences were considered significant if p<0.05). The presence of lesions in the optic nerve was assessed on double inversion recovery (DIR) MRI sequences. The comparison between the ground truth and the generated masks reported a mean (SD) Dice = 0.51(0.15). The mean T1/T2 was significantly lower in the affected eye of ON patients with detectable MRI lesions on DIR when compared to the other groups: 0.760.08 for HC (p<0.001); 0.740.006 for MS-nON (p<0.001); 0.710.12 fellow eye of ON (p<0.001); 0.740.11 affected eye of ON patients with no detectable MRI lesions on DIR (p=0.44); 0.610.09 optic nerve affected eye with lesion at MRI. Although the segmentation method yields a Dice coefficient that is not particularly high, it is acceptable given that the aim is not to measure the volume of the optic nerve, but rather to generate a mask over which to calculate the T1/T2 profile. On the other hand, when assessing small structures, even minor inaccuracies can lead to low Dice coefficients, as the metric is particularly sensitive to slight mismatches in small volumes. Validation in a cohort acquired using a scanner from a different vendor is necessary to confirm the generalizability of the proposed network. Assessment of the integrity of the optic nerve appears to be a feasible option using conventional MRI sequences. Investigation of its potential clinical applications seems warranted.
Helena SÁNCHEZ-ULLOA, Paola AJDINAJ, Neus MONGAY-OCHOA, Eugenio FUNELLI, Manel ALBERICH, Gemma PIELLA, Àlex ROVIRA, Jaume SASTRE-GARRIGA, Deborah PARETO (Barcelona, Spain)
13:30 - 15:00
#47736 - PG555 Exploring Spinal Proprio-Motor Networks and Their Plasticity Using fMRI.
PG555 Exploring Spinal Proprio-Motor Networks and Their Plasticity Using fMRI.
Muscle proprioception, conveyed through spinal and supraspinal pathways, enables movement perception and control. Proprioceptive afferents relay muscle stretch information from the muscle spindles to the spinal cord, where direct connections between Ia afferents and motor neurons form the basis of the myotatic reflex. This sense can be manipulated using muscle tendon vibration, which selectively activates primary endings of muscle spindles and induces kinesthetic illusions in the absence of actual movement [Kavounoudias et al., 2023].
While proprioceptive integration has been widely investigated at the human cerebral level using neuroimaging approaches, its spinal mechanisms remain less explored, especially using non-invasive techniques. Post-mortem studies have mapped spinal cord anatomy, while its functional properties have been explored via electrophysiology, invasive epidural stimulation, and clinical observations. In the past decade, spinal functional MRI (fMRI) has overcome key technical challenges, emerging as a powerful, non-invasive tool for studying spinal circuits [Landelle et al., 2021].
We developed a protocol combining fMRI with proprioceptive vibration to investigate the spinal proprioceptive-motor circuits. Two studies were conducted: one investigating cervical spinal cord activity with bilateral upper limb muscle stimulation (at the wrist, elbow and shoulder levels) in 24 participants; A second study explored lumbar spinal cord activity with bilateral lower limb muscle stimulation (at the ankle, knee and hip levels) in 28 participants. The second study also assessed the effects of a two-week proprioceptive training program on spinal activation patterns.
After acquiring MRI data using optimized sequences, spinal data preprocessing involved custom steps primarily using functions from the Spinal Cord Toolbox (SCT). Previously, normalization was based on vertebral landmarks, but for functional analysis, neural landmarks would seem more reliable. To improve the accuracy of overlaying individual data onto the template, we collaborated with the SCT team to develop a tool for identifying spinal rootlets, which enabled precise identification of individual spinal levels [Valosek, Mathieu, Schlienger et al, 2024]. These levels were then normalized to the PAM50 template. Additionally, we created a probabilistic map of spinal levels for our cohort, which better captures inter-participant variability. Spinal activation patterns extracted from the upper limb stimulations showed lateralization of activity towards the ipsilateral and ventral hemicord, with clear functional distribution along the cervical axis. For example, anterior deltoid activity peaked at C5, biceps at C7, and wrist flexors at C8, demonstrating a rostrocaudal gradient linked to proximodistal muscle location. Secondary dorsal activation peaks were observed, likely corresponding to tactile afferent stimulation.
Density maps showing the number of participants who activated each spinal level demonstrate that proprioceptive stimulation can engage multiple spinal levels simultaneously, with notable variability across participants. In some cases, participants activated several spinal levels during the stimulation, and this activation was often asymmetrical between the left and right sides.
In the lower limb study, the left hip showed activation at T11, the knee at L3-L4, and the ankle at L5-S1, with consistent lateralization and ventral-side dominance. After two weeks of proprioceptive training of the left knee, proprioceptive stimulation of the trained knee yielded more focal & ipsilateral activation across the lumbar axis, suggesting an effect of training on synaptic excitability of the proprio-motor circuit. This work provided detailed insights into spinal cord activity during proprioceptive stimulation, confirming the feasibility of spinal fMRI as a tool for investigating proprioceptive motor circuits. The findings align with recent literature on proprioceptive processing but also highlight significant inter-participant variability in spinal cord functional organization. The use of spinal fMRI to capture multiple activation sites along the cervical and lumbar axes offers a more comprehensive understanding of spinal activity compared to traditional methods like electrophysiology. The observed plasticity after proprioceptive training suggests that the spinal circuits underlying proprioception can be modulated by targeted interventions, which may have implications for neuroplasticity in spinal injuries or rehabilitation. In laying out a network-level map of proprioceptive-motor circuits using non-invasive fMRI, we reveal activation patterns and inter-individual variability, which challenge traditional views of spinal cord functional organization. This variability accross participants highlights the potential of spinal fMRI to offer deeper insights into human spinal functional organization and its plasticity, both in health and disease.
Raphaëlle SCHLIENGER (Marseille), Caroline LANDELLE, Sergio D HERNANDEZ-CHARPAK, Daniela M PINZON-CORREDOR, Julien SEIN, Bruno NAZARIAN, Jean-Luc ANTON, Grégoire COURTINE, Anne KAVOUNOUDIAS
13:30 - 15:00
#47723 - PG556 B₀-corrected single-shot spiral MRI of the cervical spinal cord at 7 Tesla.
PG556 B₀-corrected single-shot spiral MRI of the cervical spinal cord at 7 Tesla.
Many advanced neuroimaging techniques, such as fMRI and diffusion-weighted imaging, depend on fast acquisitions, which are most commonly achieved with single-shot EPI [1-3]. Spiral imaging offers several theoretical advantages over EPI, including shorter TE, increased robustness to flow and motion, and more efficient usage of the gradients [4,5]. However, spiral sampling is inherently sensitive to imperfections in the gradient system and B₀-field inhomogeneities, causing image blurring. While great advances have been made to address these challenges for spiral imaging of the brain ([4]), comparable progress for the spinal cord remains limited. In this work, we aim to implement high-resolution single-shot spiral imaging of the cervical spinal cord at 7 Tesla, focusing on B₀-map estimation and correction during image reconstruction.
Single-shot spiral images were acquired in a healthy volunteer on a 7T Terra MR system (Siemens Healthineers) using a 1Tx, 24Rx cervical spine coil (MRI.TOOLS). Four spirals of varying resolution and acceleration factor were acquired (parameters in Fig.1), each including 20 repetitions of a single transverse slice at C3 mid-vertebral level. For coil sensitivity- and B₀-map estimation, a dual gradient echo (GRE) scan of the same slice was performed prior to the spiral acquisitions (parameters in Fig.1).
Images were reconstructed with a CG-SENSE algorithm ([6]) using the open-source pipeline GIRFReco.jl ([7,8]), with and without B₀-correction. The trajectories were corrected with the system gradient impulse response function (GIRF) ([9,10]), measured with a phantom-based method [11]. B₀ field maps were estimated with a regularized method ([12]) using varying regularization parameters (β = 0.0005–0.05) to investigate the trade-off between noise and precision. The resulting spiral image reconstructions were visually assessed, posing particular attention to the cord region. Finally, time series of all repetitions were reconstructed using a B₀-map with β = 0.01. Mean images and maps of temporal signal-to-noise ratio (tSNR) were computed, and mean tSNR values within a spinal cord region of interest (ROI) were extracted using MRIcroGL. Uncorrected spirals acquired with low acceleration (R = 1-3) were strongly blurred and distorted by off-resonance (Fig. 2a). After B₀-correction (Fig. 2b), blurring was visibly reduced, particularly around the spinal cord and CSF. The corrected spirals with 1.08/1.00 mm resolution enabled particularly clear identifiability of anatomical features. The spiral with highest acceleration (R=4) showed minimal off-resonance effects, but suffered from noise, and B₀-correction had little effect on improving image quality.
Figure 3 shows B₀-maps estimated with varying regularization parameters together with the corresponding spiral image reconstructions. With increasing β, the B₀-profile became smoother and extreme values were reduced (black arrows). Correspondingly, spiral images reconstructed with low β were noisier. With increasing β, the pixelation was reduced and image quality increased, but at the cost of increased blurring. At β = 0.05, reconstructions became overly smooth, with reduced contrast between the spinal cord and CSF. For the spirals with 1.08/1.00 mm resolution, β = 0.01 appeared to offer a sweet spot between noisy and blurred appearance.
The mean images of the time series (Fig. 4a) showed satisfactory image quality for the spirals with R=1-3, although the lowest resolution (1.71 mm) with limited anatomical detail. The corresponding tSNR maps (Fig. 4b) revealed a mean tSNR within the spinal cord ROI of 20, 12, and 9 for the 1.71, 1.08, and 1.00 mm resolution images, respectively. For the image with 0.74 mm resolution, the mean tSNR was 11.2, but overall image quality remained suboptimal, consistent with earlier findings. This work demonstrated the feasibility of single-shot high-resolution spiral imaging of the spinal cord at 7 Tesla. Acquisitions with resolution up to 1 mm and acceleration factors of up to 3 yielded good visual image quality, and a mean tSNR of around 10. In comparison, EPI fMRI studies ([1]) reported tSNR up to 14.6 in the spinal cord. Correction for static B0 inhomogeneity in the image reconstruction was shown to be essential. The choice of regularization parameter in the field-map estimation also proved to have a strong impact on image quality.
The investigation was performed in one subject, in a single slice, and was meant as proof-of-principle. Further work is required to evaluate the robustness of the method across multiple slices and subjects, and to optimize the B0 correction. Single-shot spiral imaging has the potential to replace single-shot EPI in applications such as fMRI and diffusion-weighted imaging of the spinal cord, addressing limitations like poor image quality and SNR, particularly at 7T. The improved acquisitions could ultimately facilitate functional and microstructural investigations of the spinal cord.
Maria Leseth FØYEN (Oslo, Norway), Laura BEGHINI, Lars KASPER, Signe Johanna VANNESJÖ
13:30 - 15:00
#46000 - PG557 White matter atlas of the in vivo human spinal cord.
PG557 White matter atlas of the in vivo human spinal cord.
The current knowledge of spinal cord white matter anatomy is based on a combination of data from animal models, and physiological and histological human studies. However, it has never been described in vivo in humans. This study aims to differentiate spinal tracts and create a spinal cord white matter atlas in humans, in vivo, using diffusion tensor imaging.
High angular resolution diffusion imaging (HARDI) of the spinal cord was acquired using a 3T MRI scanner in 49 healthy subjects, with opposed-phase encoding directions. Distortion corrections were performed using the FSL software package. Diffusion tensors were reconstructed using the generalized q-sampling imaging method. The tensor components from each subject were then registered onto a reference subject’s data. The registration utilized the Symmetric Normalization transformation type, combining both global transformations and local, more complex non-linear deformations. Mutual information (MI) was employed as the optimization metric to ensure accurate alignment. The tensor fusion was achieved by averaging the transformed diffusion tensors using the Log-Euclidean metric. Regions of interest were drawn specific to each spinal projection tract. Tractography was performed using a deterministic approach with DSI Studio. The main parameters influencing the MI were grad_step, reg_iterations, aff_iterations, and aff_sampling. With the adjustments, the mean MI value increased from -0.5 (default settings) to -0.67 (optimized settings). A spinal cord white matter atlas was built: the corticospinal tract was located in the lateral funiculus at the edge of the ventral horn; the rubrospinal tract overlapped with the corticospinal tract in the lateral funiculus; the reticulospinal tract was located in the ventral funiculus; the ventral and dorsal spinocerebellar tracts overlapped in the dorsal part of the lateral funiculus and the dorso-lateral part of the dorsal funiculus; the spinothalamic tract was located around the ventral horn, while the dorsal columns were in the dorsal funiculus of the spinal cord. The process of image registration and fusion across a cohort of 49 subjects offered a robust solution to the challenge of inter-individual variability, which is a notable limitation in many existing studies. By generating a composite spinal cord model, the present study captured the common structural features across a diverse population, leading to a more generalized and reliable anatomical reference. HARDI, combined with advanced image registration and fusion techniques, offers significant advantages for the precise mapping and differentiation of spinal tracts in the in vivo human spinal cord, establishing a new standard for spinal cord mapping.
Corentin DAULEAC (Lyon), François COTTON, Frindel CAROLE
13:30 - 15:00
#47689 - PG558 Imiomics in lumbar spinal stenosis.
PG558 Imiomics in lumbar spinal stenosis.
Lumbar spinal stenosis (LSS) is a common condition affecting about 20 % of the population aged 50 to 69 years [1]. However, currently MRI findings have a low correlation to patient’s disability level [2]. This gap highlights the need for new imaging approaches that may better reflect clinical symptoms. Imiomics is an image analysis method that aligns individual MR images to a common reference, enabling detailed, voxel-wise comparison of anatomical differences between cohorts [3]. This approach may offer new insights into the structural changes associated with LSS, which can be complex and spatially distributed.
This study aimed to evaluate Imiomics as a proof of concept for identifying anatomical differences between patients with LSS and healthy controls using lumbar spine MR images.
Two datasets were used: one comprising subjects from the LSS cohort of the Norwegian Degenerative Spondylolisthesis and Spinal Stenosis (NORDSTEN) study [4] and one dataset consisting of healthy controls (77 individuals), examined at the Carlanderska Hospital in Gothenburg [5]. For each subject, a whole T1w and a T2w sagittal lumbar spine image volume was selected. From the LSS cohort two cases were selected.
All vertebrae, intervertebral discs and central spinal fluid (CSF) were segmented using TotalSpineSeg [6], a tool based on the deep learning model nnU-Net [7]. All images were masked to a convex region enclosing the vertebrae. One randomly selected healthy control was used to define a common coordinate system and all remaining images were registered to this reference using the software Deform [8, 9]. The intensity images combined with the segmentation mask of the vertebrae and intervertebral discs were layered as input channels in the registration. Coordinates along the centerline of the spinal canal at each intervertebral disc level were used for initial image alignment and for further constraints during the registration.
Since signal intensity values varied across the MR images due to differences in acquisition parameters, coil sensitivity, etc., intensity normalization was performed using the mean CSF signal intensity, followed by histogram matching to a selected reference image.
One T1w and one T2w intensity atlas to characterize normal signal distributions was generated for the registered and normalized images of the healthy controls. For each voxel, the mean intensity and standard deviation were computed. To identify signal intensity deviations in individual LSS patients, voxel-wise absolute Z-scores were calculated by comparing patient intensities to the corresponding atlas distributions. Z-scores exceeding 1.96 were considered statistically significant, indicating abnormal signal intensity. The image registration of the healthy controls yielded mean Dice scores of 0.76±0.04 (T1w) and 0.82±0.04 (T2w). The midsagittal slice of the intensity atlas characterizing normal intensity signal distributions is illustrated in Figure 1.
Two LSS cases were selected to illustrate anomaly detection. In the first case (Figure 2), a patient with severe central stenosis (Schizas grade D at L2-L3 and L3-L4), Imomics correctly highlighted known stenosis at L2-L3 in the T2w image, and epidural fat loss in the T1w image. Images were registered with Dice scores 0.80 (T1w) and 0.84 (T2w). In the second case (Figure 3), a patient with preserved epidural fat and no or minor central stenosis (Schizas grade A), Imiomics did not depict any relevant anomalies in the midsagittal plane. The patient had foraminal stenosis, which is typically not visible in this this view. Dice scores for registration were 0.77 (T1w) and 0.78 (T2w). One challenge when evaluating differences in intensities between MR systems and protocols is the lack of standardized voxel intensity values. Despite only simple intensity normalization and analysis being employed, the results demonstrate that Imiomics could be a feasible tool for detecting anomalies in lumbar spine MR images. Since Imiomics enables untargeted analysis and visualization of deviations between patient images and healthy controls, one potential clinical application could be as a decision support system for highlighting pathological changes that require more detailed analysis. Given the challenges in comparing intensity values between systems and scan protocols, future work should focus on developing more sophisticated atlases. A promising next step could be to combine Imiomics with tools from Radiomics, which enables quantitative analysis of MR images from different systems and scan protocols on a pixel-by-pixel level [10]. Imiomics demonstrates feasibility for detecting relevant changes in the lumbar spine of patients with LSS, compared to healthy controls. In the future with more refined atlases, Imiomics may serve as a valuable tool for identifying subtle pathological changes in LSS, potentially improving the correlation between MRI findings and patient-reported disability.
Alice NILSSON (Gothenburg, Sweden), Christian WALDENBERG, Erland HERMANSEN, Hanna HEBELKA, Hasan BANITALEBI, Helena BRISBY, Kari INDREKVAM, Kerstin LAGERSTRAND
13:30 - 15:00
#47707 - PG559 Multimodal MR Imaging of Brachial Plexopathy: Preoperative Diagnostic Value Across Conditions.
PG559 Multimodal MR Imaging of Brachial Plexopathy: Preoperative Diagnostic Value Across Conditions.
Brachial plexus pathology is a serious clinical concern that can result in long-term motor and sensory impairment of the upper limb. Diagnosing brachial plexopathy is challenging and requires a multidisciplinary approach involving neurologists, radiologists, neurosurgeons, and clinical researchers (1-2). Multimodal MR imaging—including MR neurography (MRN), diffusion tractography (MRT), and MR angiography (MRA) closely aligns with clinical and electrophysiological findings, helping to pinpoint lesion location, clarify the cause, and guide surgical planning. This makes it vital for effective preoperative evaluation and treatment of patients with brachial plexopathies.
Ten patients (7 males, 3 females; age range: 18–72 years) with clinically and electrophysiologically confirmed brachial plexopathy underwent MR imaging on a 3T scanner using an 18-channel phased-array body coil and a 64-channel head/neck coil. The imaging protocol included diffusion-weighted imaging with a multishell diffusion scheme (TR/TE: 5500/83 ms; 64 directions; 80 slices; in-plane resolution: 3×3 mm²; 9 b-values ranging from 0 to 950 s/mm²; scan time: 6 minutes 18 seconds) and coronal T2-weighted 3D short-term inversion recovery sampling perfection with application optimized contrast using varying flip angle evaluation (STIR-SPACE) sequences (TR/TE: 3000/2710 ms; inversion time: 230 ms; 144 slices; in-plane resolution: 1×1 mm²; scan time: 10 minutes 56 seconds). MR angiography was also performed. Diffusion data were corrected for susceptibility artifacts using reversed phase-encoding b0 volumes via TOPUP (Tiny FSL: http://github.com/frankyeh/TinyFSL) and processed through DSI Studio (http://dsi-studio.labsolver.org). Fiber reconstruction of the brachial plexus was performed using the Generalized Q-Sampling Imaging (GQI) algorithm (Generalized q-sampling imaging). MRN and volume rendering were completed using MEDINRIA (medInria). Etiologies included trauma (n=6), thoracic outlet syndrome (TOS, n=3), and inflammation (n=1). Multimodal MRI findings matched clinical and electrophysiological diagnoses in 9 out of 10 patients (90%). In traumatic cases (n=6), MRN and DTI accurately identified root avulsions and neuromas, guiding surgical decision-making. In TOS cases (n=3), hyperintensities or trunk-level abnormalities were clearly visualized; however, one scan was compromised by severe motion artifacts. The inflammatory case demonstrated C6 root hyperintensity consistent with clinical findings. Representative MRN reconstructions and corresponding tractography are hown in Figure 1. The results of our study in 10 patients with different aetiologies of brachial plexopathy highlight the crucial role that multimodal MRI techniques—specifically MRN, MRT, and MRA, can play when integrated with electromyography (EMG) and thorough clinical examination. This comprehensive diagnostic approach not only enhances the accuracy of lesion localization and characterization but also provides critical anatomical and functional information that is essential for tailored neurosurgical planning. The ability of these modalities to visualize nerve continuity, detect signal abnormalities, and assess adjacent vascular structures offers a significant advantage over conventional imaging. Our findings support the growing consensus that a multimodal diagnostic strategy should be considered a standard of care in the evaluation and management of complex peripheral nerve pathologies such as brachial plexopathies Multimodal MR imaging, including MR neurography and diffusion tractography—shows strong concordance with clinical and electrophysiological findings in brachial plexopathy. It offers crucial preoperative insights across traumatic, compressive, and inflammatory causes, enhances lesion localization, and sharpens surgical targeting, making it an essential tool for surgical planning and management.
Supported by the Ministry of Health of the Czech Republic in cooperation with the Czech Health Research Council under project No. NW 24-08-00086 and DRO (IKEM, IN 00023001).
REFERENCES
1. Ibrahim I, Škoch A, Herynek V, Humhej I, et al. Magnetic resonance tractography of the brachial plexus: step-by-step. Quant Imaging Med Surg. 2022 Sep;12(9):4488-4501.
2. Jung JY, Lin Y, Carrino JA. An Updated Review of Magnetic Resonance Neurography for Plexus Imaging. Korean J Radiol. 2023 Nov;24(11):1114-1130.
Ibrahim IBRAHIM (Prague, Czech Republic), Ivan HUMHEJ, Antonín ŠKOCH, Theodor ADLA, Vlasta FLUSSEROVA, Dana KAUTZNEROVÁ, Simona KURKOVÁ, Dominik HAVLÍČEK, Jaroslav TINTĚRA
13:30 - 15:00
#47750 - PG560 Breast MRI: comfort evaluation of supine positioning and dedicated breast holder.
PG560 Breast MRI: comfort evaluation of supine positioning and dedicated breast holder.
MRI is commonly used for breast cancer diagnosis, especially for high-risk women and cancer staging [1]. Current breast MRI performed in prone position with breast positioned in a coil, is often perceived as uncomfortable [2]. Additionally, patient preparation and correct positioning increase the exam duration and contribute to anxiety and discomfort, finally reducing patient acceptance of the procedure. To enhance patient comfort and diagnostic specificity of breast MRI, we have developed a comprehensive approach performed in a supine position and relying on a dedicated breast coil ‘BraCoil’, motion sensors and motion correction algorithms [3-6]. To further improve comfort, we also designed an elastic breast holder to maintain the breast shape without compression during MRI.
Since the success of MRI exams largely depends on the patient's cooperation, this study aimed to evaluate the comfort experienced by volunteers undergoing supine breast MRI with our technology, by collecting direct feedback after the MRI scan. Dedicated questionnaires were developed to evaluate perceptions related to the position and to the breast holder use.
11 healthy volunteers underwent breast MRI without contrast agent injection in two positions: prone and supine (Fig. 1). The order of the two examinations was alternated across participants. Before the supine MRI, each wore a personalized elastic breast holder shown in Figs. 2a & 2b, developed in collaboration with Bioserenity (France), suited to her morphology. A second supine MRI was performed afterwards without the holder. After imaging, participants completed two questionnaires: one comparing comfort, positioning, pain and anxiety between the two positions, and another assessing the breast holder's comfort, usability, and size suitability. Due to an organizational issue, one participant only completed the first questionnaire, yielding 11 responses for position comparison and 10 for holder evaluation. Investigation procedures were conducted under ethical protocol EDEN (NCT05218460).
Comfort scores for the two positions (from "very uncomfortable" to "very comfortable") were compared using a one-sided sign test. Paired binary responses regarding pain or anxiety during the examination (Yes/No) in both positions were compared using McNemar's test. A p-value < 0.05 was considered statistically significant for all analyses. Figure 3 shows the results for position comfort evaluation. The prone position was rated as moderately uncomfortable by 55.5% of the volunteers, whereas the supine position received 100% positive ratings. Overall, comfort scores were significantly higher for supine position compared to prone (p = 0.035). 82% of participants judged the supine position to be more comfortable than the prone. In terms of pain, 45.6% of volunteers reported discomfort in the prone position, compared to only 18.2% in the supine; however, this difference did not reach statistical significance (p = 0.180). On the other hand, the supine position was significantly more anxiogenic (p = 0.046), with 45.6% of participants reporting anxiety.
Results from the breast holder evaluation are shown in Fig. 4. All volunteers found the holder easy or very easy to use. For 70%, it took less than one minute to put on. The majority (80%) of participants felt psychologically comfortable wearing the holder and 20% felt slightly embarrassed. The examination with the breast holder was rated as equally comfortable by 50% of the volunteers and as more comfortable by the remaining 50%. Visual inspection of MRI images with and without the holder showed that, as expected, the holder slightly improved the similarity between the supine and prone breast shapes, particularly in women with larger breasts (Fig. 2c). Some studies showed supine MRI's feasibility for breast imaging, but women comfort has not been assessed [7]. Along with the gains in image quality and correlation with other procedures [5,8], the positive feedback from women on comfort and pain with our supine MRI protocol contributes to a higher acceptance of breast MRI, compared to prone exam, enabling its more widespread use for diagnosis of cancer. Nevertheless, anxiety is more commonly felt in the proposed supine protocol, primarily due to the view of the MRI tunnel. This can be managed by using sleep masks, mirrors or audio-visual sensory immersion devices to create a more comfortable scanning environment [9-10].
Meanwhile, the breast holder, found to be user-friendly, appears to provide only a slightly advantage in comfort. Further validation on larger cohorts and on patients with breast lesions is ongoing to allow for broader generalization of these findings. The proposed supine breast MRI approach provides better comfort than the routine position, leading to a better acceptance of breast MRI by women. Using a specific breast holder further improves patient satisfaction, but its effect on breast morphology suggests that further evaluation is necessary.
Barbara FISCHER (NANCY), Karyna ISAIEVA, Lucile BASTIEN, Sarah EL-MAGHRANI, Guillaume DROUOT, Gabriela HOSSU, Nicolas WEBER, Samuel ROGIER, Philippe HENROT, Jacques FELBLINGER
13:30 - 15:00
#47926 - PG561 Hemodynamic Forces in ST-elevation myocardial infarction - anterior versus non-anterior.
PG561 Hemodynamic Forces in ST-elevation myocardial infarction - anterior versus non-anterior.
ST-elevation myocardial infarction (STEMI) is the most severe form om myocardial infarction, and whether the ischemic damage is localized anterior or inferior (e.g. non-anterior) can affect the prognosis. Ventricular remodelling may occur due to and scarring and fibroisis which affects all cardiac healing process and causes permanent changes in cardiac shape, geometry, size and function e.g. adverse remodelling. The cardiac magnetic resonance imaging (CMR) software tool Hemodynamic Forces enable retrospective analysis of the intracavitary gradient pressures direction during a heart cycle, a measurement of cardiac efficiency. A few studies have been published using this software, but none has compared patients with anterior and non-anterior STEMI with as high timely resolution as in this study. The purpose with this study is to visualize different patterns of the hemodynamic force after anterior and non-anterior STEMI over time.
This exploratory study was based in the Stunning in Takotsubo versus Acute Myocardial Infarction cohort (NCT04448639). During analyzation qualitatively, different pathological patterns appeared, Three patients with anterior STEMI and three patients with non-anterior STEMI with no history of cardiac disease were examined with CMR imaging at three time points after hospitalization, the acute , subacute, and chronic) phases. Based on CINE imaging, calculations of the hemodynamic forces proceed from endocardial border throughout the heart cycle, average mitral valve diameter 4, 2 and 3 chamber view and the aortic valve, 3ch (Fig. 1). 6 patients were included in the study based on infarct localization, presence of adverse remodelling, sex, and age and presentation of different longitudinal patterns from a larger cohort. Divided into anterior (A1, A2, A3) vs non-anterior (nA1, nA2, nA3), two males, one female each, two had no adverse remodelling, one female also one male (Fig. 2). In these few cases the curves show difference in appearance whether the infarcts are anterior or non-anterior despite infarction size (Figs. 3-4). Both A3 and nA3 had no adverse remodelling, cardiac function recovered, the appearance of the curves is lower in the acute phase and increase in force over time (Fig. 3). Plotting the peak value of force over time, the trend indicates that in these cases increased or preserved force led to adverse remodelling whilst a reduced initial force eventually recovers. Our results show differences in the appearance of both anterior and non-anterior STEMI patients. For non-anterior STEMI, the time curve of the hemodynamic force may appear abnormal in the acute phase then recover until the subacute phase, in contrast to anterior STEMI were the hemodynamic curve have a normal appearance in acute phase but not the subacute. Similarly, for both of those with no remodelling the general peak force are lower in acute phase that accumulates over time. Is it posslble that a down regulation of the longitudinal forces during the systolic peak may affect the recovery and prevents adverse remodelling after STEMI, could changes in hemodynamic forces over time have a prognostic value in STEMI patients. In present study we have identified different patterns of the hemodynamic forces depending on localization, anterior or non-anterior myocardial infarction also outcome adverse remodelling or not. These are only a few cases not enough to decide on any significance and a larger sample is necessary and further evaluation.
Christina PETTERSSON (Gothenburg, Sweden, Sweden), Björn REDFORS, Andersson AXEL, Christian L POLTE, Kerstin LAGERSTRAND
13:30 - 15:00
PG561 Hemodynamic Forces in ST-elevation myocardial infarction - anterior versus non-anterior.
Christina PETTERSSON (PhD student) (Poster Displayed, Gothenburg, Sweden, Sweden)
13:30 - 15:00
#47861 - PG562 Uncovering the role of superior colliculus in rats dynamic vision with BOLD and ADC-fMRI.
PG562 Uncovering the role of superior colliculus in rats dynamic vision with BOLD and ADC-fMRI.
In the mammal brain, the perception of a recurrent visual stimulus involves both the primary visual cortex (V1) and the superior colliculus (SC). In rats, stimuli at a higher frequency than the Flicker Fusion Frequency (FFF) threshold led to continuity illusion and showed a specific pattern of positive and negative BOLD-fMRI responses [1-2]. To better understand the functional mechanisms behind this transition from static to dynamic vision, we compared the fMRI response to visual stimuli below and above the FFF threshold and investigated the effect of sex and magnetic field strengths. We also compared BOLD-fMRI to apparent diffusion coefficient (ADC)-fMRI responses[3-5], which relies on neuromorphological[6-7] rather than neurovascular coupling.
Twelve Sprague-Dawley rats (n=6 females) were scanned on a 14T Bruker MRI system with a volume transmit coil and a receive-only surface coil, and 6 additional females were scanned on a 9.4T Bruker MRI system with a volume transmit coil and a receive-only cryoprobe. Table 1 presents the acquisition parameters of the T2-weighted anatomical scan and of BOLD and ADC-fMRI timeseries (isotropic spherical diffusion encoding with alternating b-values of 200 and 1000 s/mm²). Rats were sedated using medetomidine following initial anaesthesia with isoflurane. fMRI acquisitions started 30 min after stopping isoflurane and consisted in blocks of 16s of stimulation (blue light flickering at a frequency of 1Hz or 25Hz) and 24s of rest, repeated 12 times. Two runs were acquired per frequency and per functional contrast.
fMRI images were denoised[8] and corrected for Gibbs ringing[9], distortions[10], and motion[11]. ADC-fMRI timeseries were computed from the co-registered b200 and b1000 timeseries. Single-level GLM with boxcar function was performed on BOLD-fMRI and results were registered to a template for group-level GLM. To reduce assumptions on ADC-fMRI response, first-level GLM was performed with a boxcar function convolved with Finite Impulse Response (FIR)[12]. Responding voxels were pooled across rats and classified using a K-means clustering algorithm. At 14T, in response to 1Hz stimulation, both sexes showed overwhelming bilateral positive BOLD response in the SC and V1 (Fig.1). In contrast, 25Hz visual stimulation elicited a negative BOLD response in V1 of all animals, and in the lateral SC of males. Plotting the average responses only showed a difference between males and females in the amplitude of the response to 25Hz stimulation in the SC.
In females, the BOLD-fMRI activation maps and response shapes were overall very similar across different gradient strengths, although the signal amplitude was consistently lower at 9.4T (Fig 2). An exception can be noted for SC at 25Hz, as two peaks can be noticed at 14T only (Fig. 2G).
ADC-fMRI response was very similar between 1Hz and 25Hz stimulation in females at 9.4T (Fig. 3). Clusters of voxels showing a negative ADC response were found in the medial part of the SC, in the hippocampus, and in the corpus callosum, in regions where BOLD response was positive. A positive ADC response, very similar to the BOLD one, was found in the lateral part of the SC. At 25Hz, we reproduced the pattern observed by Gil et al. showing positive BOLD in the SC and negative BOLD in V1 [1-2]. In addition, our results highlight a difference between males and females, showing negative BOLD in the lateral SC in males.
The activation maps derived from 14T and 9.4T MRI mainly showed an expected scaling of BOLD amplitude with field strength. In the SC at 25Hz, the shape of the responses differed at 9.4T, which may be attributed to the increase in small-vs-large vessel relative contributions at lower fields[13-14] .
ADC-fMRI is a promising complementary approach to probe brain function independently of the hemodynamic response[15]. Interestingly, ADC-fMRI response showed opposite signs between the lateral and medial SC, even with 1Hz stimulation. In the white matter and in the medial SC, negative ADC response was measured in the presence of positive BOLD suggesting a neuromorphologically-driven contrast. However, in the lateral SC, the positive ADC response was very similar to BOLD and probably due to residual magnetic susceptibility contributions to the ADC timecourse. We confirmed the key role of the SC in the visual perception of high frequency stimuli. The differences spotted in BOLD fMRI across sex and field strength suggested a separation between medial and lateral SC in the complex excitatory/inhibitory observed pattern. Using ADC-fMRI to probe brain function without vascular coupling could help better understand interactions between regions of the visual network and the white matter connecting them.
Jean-Baptiste PEROT (Lausanne, Switzerland), Andreea HERTANU, Arthur SPENCER, Jasmine NGUYEN-DUC, Nikolaos MOLOCHIDIS, Valerio ZERBI, Maxime YON, Ileana JELESCU
13:30 - 15:00
PG562 Uncovering the role of superior colliculus in rats dynamic vision with BOLD and ADC-fMRI.
Jean-Baptiste PEROT (Postdoc) (Poster Displayed, Lausanne, Switzerland)
13:30 - 15:00
#47713 - PG563 Reproducible detection of fine-grained face selective patches in the human prefrontal cortex using high resolution fMRI at 9.4 Tesla.
PG563 Reproducible detection of fine-grained face selective patches in the human prefrontal cortex using high resolution fMRI at 9.4 Tesla.
The prefrontal cortex (PFC) is commonly associated with executive functions such as planning and working memory[1, 2]. And yet, both monkey and human PFC have been shown to contain regions that selectively respond to visual stimuli of faces versus other objects[3, 4, 5]. Recent studies using high-resolution fMRI in macaques revealed that these ”prefrontal face-patches” make up a small portion of the prefrontal cortex and possibly part of a larger area of fine-grained mosaic of object-specific patches[6, 7], similar to what has been found in the inferotemporal cortex[8, 9, 10]. Currently, there is no strong evidence of such a fine-grained topographic organization of visual objects in human PFC. We hypothesize that evidence of such a topographic organization in human PFC is washed out by a combination of population averaging and low resolution imaging. In this work, we used high-resolution fMRI at 9.4 Tesla, to investigate if fine grained face patches can be detected reproducibly in the PFC of individual humans.
Five subjects completed three scan sessions each. All participants provided informed consent prior to each scan, in compliance with local IRB regulations. The stimuli contained images of faces[11] and manipulable inanimate objects (MIO) including tools and food[12]. A counterbalanced block design was used (Fig. 1a). A one-back task was used to control attention (Fig. 1b).
All functional data was collected at 9.4 Tesla (Magnetom Siemens, Erlangen, Germany) using the AC84 head gradient (333 T m−1s−1, 80 T m−1). The coil consisted of a 16-channel dual-row transmit array, with a 31-channel receiver array insert[13]. Functional data was collected using a slab-selective, three-dimensional echo-planar imaging (3D-EPI) sequence[14]. A 52-mm thick slab was placed true axial, just above the orbital gyrus, with a 192-mm square in-plane. The resolution was 0.75 mm isotropic. A 3-s volume TR and 19-ms TE were achieved using a 3 × 2 undersampling factor, with a CAIPI shift of 1 in the kz-direction and a shot segmentation factor of 2[14, 15].
Preprocessing was performed using AFNI[16]. GLM analyses were performed with and without smoothing (1.5 mm). The first 3 volumes of each run were removed. The average task performance across subjects was 92±9%.
Figure 2 shows the unsmoothed GLM result of the 2nd session of subject 1. Similar to previous reports[5], the areas responding strongly to faces were located around the inferior frontal sulcus in the right PFC. Interestingly, our face patches were much smaller than previously reported, probably due to the significant improvement in resolution (3.0 mm vs 0.75 mm iso). Notably, we also observed small patches that appear to show signs of selectivity towards images of MIO nearby.
Figure 3 shows the activation pattern observed in both unsmoothed and smoothed results from all 3 sessions of subject 5. In this subject, both face- and MIO-clusters were observed in consistent locations throughout all sessions.
Figure 4 shows smoothed results measured in all other subjects. The 3rd session of subject 1 and the 1st session of subject 4 showed weak or no evidence of face-selective activation in the right PFC. The location of the face patches in subject 1 appeared to be lower in the brain than the others. This is likely a visual effect due to a difference in head tilt. Relative to the face patches, the location of MIO patches differed considerably between individuals. In some subjects, MIO patches tended to lie near the sulcus, whereas in others they were closer to the gyrus. In the 2nd session of subject 3, MIO patches were found in both of these locations. Yet, session 1 only showed the sulcal patch, whereas the 3rd session only showed the gyral patch. It is likely that these face- and MIO-selective cortical patches are not in the exact same spot across subjects in MNI space. Therefore, averaging across subjects could give the impression that these face-patches are larger, possibly overshadowing neighboring patches with specific selectivity for other well-defined features. To further investigate this hypothesis, we plan to coregister the data to T1-weighted anatomical images and conduct surface-based analysis.
Future work should also consider expanding the range of object categories as well as separating tools and food into separate categories. This could allow a more complete mapping of the topographic organization in the PFC, as well as further validate the specificity of these patches with regards to other types of objects. Our results indicate that face-selective cortical patches in the human PFC are very small, and can be reproducibly observed across sessions. In some subjects, MIO-selective patches were found nearby, suggesting that some additional topographic organization of objects may exist in the human PFC. More generally, these preliminary results highlight that, albeit challenging, reproducible high-resolution fMRI of category sensitivity in the human PFC is possible at 9.4 T.
Noriya ASAMI (Nijmegen, Japan), Desmond H. Y. TSE, Yoichi MIYAWAKI, Logan DOWDLE, Benedikt A. POSER, Wim VANDUFFEL, Timo VAN KERKOERLE, Martijn A. CLOOS
13:30 - 15:00
#47866 - PG564 Towards Layer-fMRI of the Human Insula During Pain Perception.
PG564 Towards Layer-fMRI of the Human Insula During Pain Perception.
The insular cortex, deeply folded within the lateral sulcus, has been repeatedly implicated as a central hub for pain processing in humans (Horing & Büchel, 2022; Segerdahl et al., 2015). Functional imaging studies suggest a gradient along its anterior–posterior axis: anterior regions encode cognitive-affective components such as pain anticipation, while posterior regions more closely track stimulus intensity. This functional axis aligns with cytoarchitectonic subdivisions observed in both humans and non-human primates (Cerliani et al., 2012), where different insular subregions exhibit distinct functional connectivity profiles. The dorsal posterior insula, in particular, receives lamina I spinothalamic input and serves as a primary cortical target for nociceptive afferents (Craig, 2009).
To bridge the gap between insights from animal models and macroscopic human neuroimaging, recent advances in high-resolution fMRI allow investigation of cortical processing at the mesoscopic level—across cortical layers—enabling directional inferences about feedforward and feedback signaling. However, applying layer-fMRI to the insula remains technically challenging due to its curvature, depth, susceptibility to signal dropout, partial volume effects, and pronounced physiological noise.
Here, we present the development of a well-powered study design with a MR protocol optimized for laminar fMRI of the insula during pain perception.
Participants received repeated thermal stimuli (48.5°C, 4.6 s plateau, 70°C/s ramp rate) on the left forearm using a CHEPS thermode connected to a TSA-2 system (Medoc). Each session consisted of 40 heat stimuli, with half presented as predictable—announced by a brief cue—and half as unpredictable.
Seven healthy subjects were scanned on a Siemens Prisma 3T system equipped with a 64-channel receive head coil. Each subject participated in six scanning sessions across two separate days. Functional imaging was performed using a 3D gradient-echo EPI (GE-EPI) BOLD sequence (Stirnberger & Stöcker, 2021) with the following parameters: voxel size = 0.82 mm isotropic, TR = 2230 ms, TE = 28 ms, partial Fourier = 6/8, 26 sagittal slices. In addition, a SS-SI-VASO sequence with the same readout and resolution was used to generate T1-weighted images exhibiting the same distortions as the functional images. High-resolution anatomical reference images were acquired using a MP2RAGE sequence with matched voxel size.
For the first three participants, both BOLD and VASO sequences were acquired to compare temporal signal-to-noise ratio (tSNR) and contrast-to-noise ratio (CNR). Based on these comparisons, subsequent sessions employed only the BOLD sequence for task-based fMRI, while the VASO sequence was retained for a short resting-state acquisition to generate distortion-matched T1-weighted images.
All functional (BOLD) and resting-state (VASO) images were denoised using NORDIC (Vizioli et al., 2021) to reduce thermal noise and then motion-corrected in SPM12 using a spatial weighting mask of the insular cortex. Mean images of VASO series were used to compute a distortion-matched T1-weighted image. Co-registration was performed in two steps: first, the VASO-derived T1 image was linearly aligned to the mean BOLD image; second, the MP2RAGE anatomical image was non-linearly aligned to the VASO T1-weighted image. First-level GLMs were computed in SPM12, incorporating paradigm regressors, 24 motion parameters, and RETROICOR physiological noise regressors, all convolved with a canonical HRF. In the first three subjects, we compared tSNR and CNR between four sessions (160 trials) of BOLD and VASO acquisitions. Across all subjects and both metrics, the BOLD time series outperformed VASO. Furthermore, activation maps based on the VASO sequence showed strong signal from large arteries, particularly the middle cerebral artery. Based on these findings, all subsequent sessions were conducted using the 3D GE-EPI BOLD sequence.
In all seven subjects, the strongest activation for the pain contrast was observed in the dorsal posterior insular cortex or in an adjacent opercular region. This spatial pattern is consistent with the location of nociceptive input reported in macaque tracer studies (Craig, 2009). In four subjects, the contrast between predictable and unpredictable pain yielded significant activation, with foci located more anteriorly. However, these activations were less spatially consistent across subjects than the main pain contrast. We established a robust and high-resolution (0.82 mm) protocol for investigating pain-related activity in the human insula using 3T fMRI. Our preliminary results are in line with anatomical findings from non-human primate studies, supporting the dorsal posterior insula as a key target for nociceptive input. In the next phase, we will analyze these data at the laminar level to assess feedforward (nociceptive input) and feedback (predictability modulation) processes within the insular cortex.
Ole GOLTERMANN (Hamburg, Germany), Christian BUECHEL
13:30 - 15:00
PG564 Towards Layer-fMRI of the Human Insula During Pain Perception.
Ole GOLTERMANN (Poster Displayed, Hamburg, Germany)
13:30 - 15:00
#47791 - PG565 Preliminary Evidence that Cold-Induced Pain Disrupts Functional Connectivity in Knee Osteoarthritis Patients with Chronic Pain: An rs-fMRI Study.
PG565 Preliminary Evidence that Cold-Induced Pain Disrupts Functional Connectivity in Knee Osteoarthritis Patients with Chronic Pain: An rs-fMRI Study.
Chronic pain in knee osteoarthritis (KOA) is associated with central nervous system changes, particularly in patients exhibiting widespread, nociplastic pain features. While resting-state fMRI (rs-fMRI) can detect intrinsic brain alterations, the effects of acute pain stimulation on functional connectivity remain underexplored. To address this, we employed the cold pressor gel test a validated, safe, standardized, and short-duration method for inducing acute pain (1,2) by applying cold gel to the non-dominant hand during scanning. This protocol allows for controlled investigation of pain-evoked neural responses. We hypothesized that patients with widespread nociplastic pain will exhibit greater disruptions in pain-related networks (e.g., thalamocortical, salience, sensorimotor) during cold stimulation, reflecting enhanced central sensitization and neuroplastic alterations.
Four patients with KOA and widespread pain (3 females, 1 male; mean age = 59.75 years) participated in the study. Each underwent MRI scanning on a 3T GE system, which included high-resolution anatomical and functional BOLD imaging. fMRI scanning consisted of three 8-minute fMRI sessions: resting state baseline (Rest), cold stimulation (ICE), and recovery. The cold pressor gel test involved applying cold gel to the non-dominant hand for two minutes during the ICE session. Preprocessing was performed using the CONN toolbox, including fieldmap-based realignment, slice-timing correction, normalization to MNI space, smoothing (8 mm FWHM), and CompCor-based denoising. Seed-based connectivity analyses were performed using a weighted-GLM on Fisher-transformed correlations. Seeds included bilateral thalamus, posterior cingulate cortex (PCC), anterior cingulate cortex (ACC), anterior insula, sensorimotor and visual cortices, brainstem, ICE vs. Rest contrasts were assessed at voxel-level p < 0.001 with cluster-level FDR correction (p-FDR < 0.05). All analyses were exploratory and interpreted cautiously due to the limited degrees of freedom. ICE stimulation significantly effected connectivity between key seed regions and specific target brain regions involved in pain processing. Posterior cingulate seed connectivity decreased with the right occipital pole (T = –46.40; 21 voxels; p-FDR = 0.00022) (Fig.1 (a)). Anterior cingulate seed showed reduced connectivity with multiple regions. The most significant decrease was observed in the orbitofrontal cortex (T = –106.75; 18 voxels; p-FDR = 0.00014). Sensorimotor cortex seed connectivity declined with bilateral M1/S1 (e.g., T = –50.45, x = –22, –88, –18; 15 voxels; p-FDR = 0.00034) (Fig.1 (b)). Right thalamus showed increased connectivity with frontal pole (T = +38.41; 16 voxels; p-FDR = 0.00047), while the left thalamus and brainstem showed decreased connectivity with occipital and cingulate regions. All findings were thresholded at voxel-level p < 0.001 and cluster-level p-FDR < 0.05. These preliminary results highlight how acute cold stimulation modulates (1,2) functional connectivity between key brain regions involved in pain processing, including thalamic, salience, sensorimotor, and brainstem networks. The observed alterations, particularly in thalamocortical and salience-related connectivity, may reflect underlying mechanisms of central sensitization and nociplastic pain in patients with KOA. We are continuing to recruit additional participants to further validate and expand upon these findings.
Mahnaz TAJIK (Hamilton, Canada), Bhanu SHARMA, Dinesh KUMBHARE, Michael D NOSEWORTHY
13:30 - 15:00
#47334 - PG566 Volumetric Study of Substantia Nigra on 3T MRI across different Movement Disorders.
PG566 Volumetric Study of Substantia Nigra on 3T MRI across different Movement Disorders.
Recent advances in magnetic resonance imaging (MRI) techniques have improved the evaluation of the substantia nigra (SN), a millimeter-sized structure in the midbrain that plays a fundamental role in modulating movement and reward functions¹. MRI studies have shown that, in patients with movement disorders, SN degeneration is characterized by loss of T2 hyperintensity signal, particularly in the dorsal region known as the nigrosome 1, also referred to as the “swallow tail sign”². This study aims to assess SN volume on Susceptibility Weighted Imaging (SWI) in healthy controls (HC) and in patients with normal pressure hydrocephalus (NPH), Parkinson's disease (PD), adult-onset Huntington's disease (HD), and juvenile-onset Huntington's disease (jHD).
SWI allows visualization of magnetic susceptibility changes due to iron accumulation following SN degeneration³ (Figure 1). The MRI protocol used in this study was acquired on a hybrid 3T PET-MRI scanner (Biograph mMR, Siemens Healthineers, Forchheim, Germany), including an SWI sequence targeting the mesencephalic region, optimally angled for better visualization of the SN (voxel size of 0.7x0.7x1.2 mm, TR (repetition time) = 29 ms, and TE (echo time) = 18 ms. Manual segmentation was performed using MRIcron software to obtain two volumes of interest (VOI), one for right SN(Figure 2). To limit the influence of individual head size and presence of artifacts, the three most representative slices were segmented for each subject. The analysis included 5 patients with normal pressure hydrocephalus (NPH), 7 patients with Parkinson's disease (PD), 6 patients with Huntington's disease (HD), 3 with juvenile Huntington's disease (jHD), and 10 healthy controls (HC). The collected data showed the following average volumes and standard deviations for each volume: HC = 0.41 ±0.059 cm³; NPH = 0.35 ±0.068 cm³; PD = 0.34 ±0.066 cm³; HD = 0.33 ±0.059 cm³; jHD = 0.23 ±0.059 cm³ (Figure 4). The analysis indicates that the VOIs are larger in the HC compared to the other groups, with a particularly noticeable gap when comparing to patients with jHD. In PD, the VOIs are lower than those in the HD, NPH, and HC groups; however, these differences are not statistically significant. To assess statistical significance, a two-sample t-test was conducted with a threshold p-value<0.05. In table shown in Figure 5, the statistically significant values are highlighted in yellow. The following observations were made regarding the differences between right and left VOIs: between HC and PD, only the left VOI and total VOI show a significant difference; between HC and HD (both adult and juvenile), all differences in VOIs are significant; between HC and NPH, there are no significant differences; between PD and HD, all VOI differences are significant, while there are no significant differences between PD and NPH; between HD and jHD, only the right VOI and total VOI are significantly different, while the left VOI does not show a significant difference; and between jHD and NPH, all VOI differences are significant. Various studies confirm primary damage to the substantia nigra in Parkinson's disease, characterized by the death of dopaminergic neurons, which leads to iron release and perpetuates the damage⁴. This results in movement disorders that can initially be treated with pharmacological therapies⁵, but ultimately compromise the patient’s quality of life. However, Parkinson's disease is not the only condition associated with movement disorders. In normal pressure hydrocephalus, these disorders are not due to primary alterations in the substantia nigra but rather to a down-regulation of dopaminergic transmitters⁶. In Huntington's disease, particularly in juvenile-onset cases, more pronounced parkinsonian symptoms are observed. Overall, this study highlighted that the disorders causing primary involvement of the substantia nigra exhibited reduced volumes compared to healthy subjects. Patients with NPH showed volumes not significantly different from those in the PD and HD groups or healthy controls, likely because the mechanism in normal pressure hydrocephalus does not induce neuronal loss. The jHD group showed a marked volumetric reduction of the substantia nigra compared to all other groups, indicating significant involvement of this structure in the disease's pathogenesis. The results support the validity of assessing the volumetric alterations of the substantia nigra using SWI sequences, allowing for a more accurate characterization of various movement disorders. However, the study has some limitations, including the difference in age across groups and the limited sample size (both determined by the rarity of certain conditions and their clinical definition in terms of disease onset). It would be interesting to expand the case series by including additional disorders that cause movement disturbances and to evaluate the potential effects of therapies on the SN.
Ilaria CHIMENTO (Catanzaro, Italy), Mariaeugenia CALIGIURI, Emanuele TINELLI, Andrea QUATTRONE, Ferdinando SQUITIERI, Aldo QUATTRONE, Umberto SABATINI
13:30 - 15:00
#48020 - PG567 Definition of a Nigrosome-1 template for characterization of Parkinson’s disease at 3T.
PG567 Definition of a Nigrosome-1 template for characterization of Parkinson’s disease at 3T.
Parkinson’s disease (PD) is characterized by degeneration of dopaminergic neurons in the substantia nigra pars compacta (SNc). Within SNc, Nigrosome-1 (N1) is the first and most severely affected region in PD, with up to 98% of neuromelanin-rich dopaminergic neurons lost by time of diagnosis. In healthy brains, N1 appears as a hyperintense region surrounded by hypointense areas. This visual pattern is often described as the “swallow-tail” sign and can be identified in the dorsolateral aspect of the SN. In PD, this sign tends to be altered as N1 degenerates, making it a potential imaging biomarker for early diagnosis. However, identifying N1 on MRI, especially at fields lower than 7T, presents several technical and anatomical challenges that can affect diagnostic accuracy: a) small size and deep location in the brain, b) irregular, often asymmetric shape, c) loss of contrast due to artifacts or pathology. In this work, we exploited optimized N1 identification on susceptibility-weighted imaging (SWI) and registration between different acquisitions schemes to derive a N1 probability map to characterize susceptibility properties of the N1 region, and assess correlates of iron accumulation also in scans where the structure is not visible.
Forty-three patients with PD and 35 healthy controls (HC) underwent 3T brain MRI (Biograph mMR, Siemens Healthcare, Forchheim, Germany) using a 16-channel PET-transparent head/neck coil. The protocol comprised i) Three-dimensional T1-weighted magnetization-prepared rapid acquisition gradient-echo sequence (MPRAGE, 176 slices, 256×247mm2 field of view, voxel-size 1×1×1mm3, TR/TE/TI=2300/2.34/900 ms, flip-angle 8°, TA=5′12″); ii) susceptibility-weighted imaging (SWI, 56 transverse planes centered on the midbrain, voxel size 0.7×0.7×1.2mm3, TR=50 ms, 5 TEs=5.88/13.62/21.62/29.62/37.96 ms, 220x213 mm2 FOV, TA=6:23); iii) a second SWI acquisition to optimize N1 visualization using TR=29 ms, TE=18ms and same voxel-size and FOV. Manual segmentations of visible N1 were performed for all HC and 37 PD patients (26 unilateral, 11 bilateral) by two expert raters and a consensus mask was created. QSM data was processed as previously described (1). Both SWI and QSM scans of each subject were non-linearly registered to corresponding T1 scan, in turn coregistered to a symmetric study-specific atlas. All N1 manual segmentations were subsequently aligned onto the atlas, thus allowing creation of a symmetric N1 probability map. Regional χ values were extracted from N1: a) using the manually segmented masks of each individual subject, i.e., only were N1 was visible on SWI; and b) coregistering the N1 probability map in each subject’s space, thus extracting one value per structure independent of its visibility on SWI. Coregistration of HC manual segmentations in a common space allowed for the creation of a probability map of a voxel belonging to the nigrosome region. Automatic and manual segmentations were highly in agreement in terms of location of the volume. Volumes on SWI scans and χ values from the QSM maps were automatically extracted using fslutils and compared between groups (Figure 1). In figure 2, we separate visible and non-visible nigrosome category, observing that results were highly comparable to manual segmentation for what concerns visible N1s. Moreover, the use of the atlas allowed us to extract χ values from regions were N1 is knowingly damaged, confirming higher QSM values than both visible HC and visible PD, suggesting greater iron accumulation and thus significant structural degradation. Quantitatively characterizing N1 in PD and its mimics is of crucial relevance, especially in the early stages of the disease. Thanks to our efforts in optimizing the alingment between structural, susceptibility-weighted, and manually segmented images allowed us to extracting one value per structure, independent of its visibility on SWI. This has several potential application and development, such as it’s use to evaluate subjects with high-risk of PD or parkinsonian syndromes.
Angelina CATRAMBONE (Catanzaro, Italy, Italy), Ilaria CHIMENTO, Maria Celeste BONACCI, Emma BIONDETTI, Iolanda BUONOCORE, Umbero SABATINI, Aldo QUATTRONE, Andrea QUATTRONE, Maria Eugenia CALIGIURI
|
Poster hall |
13:45 |
"Saturday 11 October"
Added to your list of favorites
Deleted from your list of favorites
A34
13:45 - 14:45
FT1 Oral - New MRI acquisition technology
13:45 - 13:55
#47634 - PG043 In vivo R2* and Quantitative Susceptibility Mapping on the 11.7T whole-body Iseult MRI System using Universal Pulses Transmission and Virtual Coil Reconstruction.
PG043 In vivo R2* and Quantitative Susceptibility Mapping on the 11.7T whole-body Iseult MRI System using Universal Pulses Transmission and Virtual Coil Reconstruction.
QSM at ultra-high field presents strong benefits due to an enhancement of the susceptibility effect [1]. However, B0 and B1+ field inhomogeneities make full exploitation difficult. Here we present the first in vivo R2* and magnetic susceptibility (QSM) maps obtained on a cohort of healthy subjects (PREMS cohort) acquired with the whole-body 11.7T Iseult magnet using universal pulses (UP) [2] and a virtual coil approach for coil combination [3]. Normative values of R2* and susceptibility (QSM) were derived in the basal ganglia using a combined manual and deep learning approach for tissue segmentation.
The in vivo protocol was approved by the national ethics committee and regulatory authorities (ANSM). Images were acquired in parallel transmission (pTx) mode using the UP for flip angle homogenization on nine healthy volunteers (mean age was 22.3 +/- 6.5 years old, 7 women /2 men) with no known neurological diseases. Data were acquired using the 11.7T Magnetom Iseult MRI with Siemens Healthineers VE12U software. A custom 8/32-channel pTx head coil was used for signal emission and reception [4]. Images were acquired using a whole-brain 3D Multi Echo Gradient Echo sequence with an isotropic resolution of 0.8 mm. Parameters were: FOV= 220x220x179 mm3, matrix size= 276x276x224, TR = 25 ms, TEs ranging from 3.50 to 17.36 ms with a ΔTE of 4.62 ms (4 echoes acquired), readout bandwidth = 430 Hz/pix. Flip angle was 10° and only one average was acquired. A 2x2 GRAPPA scheme and elliptical sampling were used to speed up the acquisition, leading to a scan time of 5 minutes 14 s. Raw data was saved for further processing. Magnitude and Phase images were generated offline, and coil combination was performed using a Virtual Coil approach using Matlab. Images of multiple echoes were combined using a root mean square along time dimension, thus providing both high SNR and T2*-contrasted images. Brain mask was obtained using “bet” (FSL). Transversal relaxation rate (R2*) was evaluated using a nonlinear fitting method in Matlab. Quantitative Susceptibility Mapping (QSM) images were reconstructed using several functions from the MEDI Toolbox [5-6] for both background field removal and dipole inversion, using respectively Laplacian Boundary Value (LBV) and a GPU modified L1-MEDI. Three templates of Magnitude, R2* and QSM were generated using Ants multivariate [7]. Regions of Interest (ROIs) were defined on the magnitude template using a deep learning model trained on synthetic data [8]. ROIs were then manually edited using 3D Slicer. Finally, ROI were back-projected to the subject space to provide subject-specific ROIs. Figure 1 presents the images obtained in one of the volunteers using the proposed framework for anatomical T2*w, fitted phase image, miP-SWI, R2* and quantitative susceptibility maps. Virtual coil combination provided naturally unbiased and usable magnitude images (no normalization used). Moreover, no clear evidence of abnormal signal inhomogeneity due to bad RF shimming or coil combination were identified on these images. Figure 2 presents the defined ROIs overprinted on the magnitude template, as well as the R2* and QSM templates. R2* and susceptibility values are reported in Table 1 (Image 3) for Caudate Nuclei, Putamen, Globus Pallidus, Red Nuclei and Substantia Nigra. Interestingly, several phase singularities were detected in phase images before coil combination but did not interfere on the final computed susceptibility maps (not shown). We report in this work the first R2* and susceptibility maps obtained at 11.7T on healthy subjects at 800 μm isotropic spatial resolution obtained in roughly 5 minutes. Universal pulses and virtual coil combination allowed to provide in vivo R2* and susceptibility maps without severe RF field inhomogeneity artefacts. The obtained R2* at 11.7T in basal ganglia subregions was approximately 2 times superior to the values reported in the literature at 3T [9]. Surprisingly the R2* values were two times lower than expected [10]. We now intend to compare the values obtained in the basal ganglia at 11.7T to those obtained at 3 and 7T using an age-matched cohort. We will also refine the MR acquisition protocol at 11.7T to enhance the image resolution to detect small details and that might not be visible at conventional field strength in vivo.
Mathieu SANTIN (Paris), Mélanie DIDIER, Franck MAUCONDUIT, Aurélien MASSIRE, Caroline LE STER, Romain VALABREGUE, Vincent GRAS, Michel LUONG, Alexis AMADON, Michel BOTTLAENDER, Nicolas BOULANT, Alexandre VIGNAUD
13:55 - 14:05
#46603 - PG044 Direct MRI of hard tissue in teeth.
PG044 Direct MRI of hard tissue in teeth.
Dental caries, the deterioration of the hard tissues (dental and enamel) in teeth, is the most pervasive noncommunicable disease and is a significant public health issue[1].
The growing interest in MRI as an ionizing radiation-free modality may serve as an alternative for x-ray imaging in dentistry[2].
Short-T2 methods have already been implemented to image the teeth, conveying bound water in enamel and dentin, which exhibits T2s on the order of 100s of µs[3-10]. However, MR signals of collagen (in dentin) and hydroxyapatite (in both dentin and enamel) have T2s on the order of 10µs[5, 6] and are yet to be imaged directly.
Here, we report the use of custom short-T2 methodology and hardware[11, 12] to directly image and study the ultra-short T2 components in teeth. The free induction decay (FID) signal of a healthy human tooth specimen is analyzed. The same specimen, as well as a tooth with a dental filling, are subsequently imaged using a multi-echo acquisition to observe the signal behavior of hard tissues. Image subtraction between early echo times (TEs) is performed to isolate the shortest-living components. Finally, the approach is applied in vivo.
Two human teeth specimens were acquired from a local dentist and stored in a dry environment at room temperature prior to scanning. One specimen has no apparent pathologies or fillings and is considered a healthy tooth. The other tooth has a dental filling made of a resin composite.
A high-performance insert gradient, with a strength up to 220mT/m at full duty cycle[13], and fast transmit/receive (T/R) switches[14] were deployed on a 3T Philips Achieva system. Tooth specimens were imaged using a 40mm diameter, proton-free T/R loop coil. In vivo imaging was performed using a proton-free T/R quadrature birdcage coil[15].
An FID of the healthy tooth was acquired to observe and characterize the signal behavior. Specimens and the in vivo case were imaged using the short-T2 PETRA pulse sequence[16]. For the specimens, a multi-TE protocol was used to capture the signal decay of the teeth with spatial localization. TE for PETRA is defined as TE = DT + 1/(2BW), with DT and BW being the dead time and imaging bandwidth, respectively[17]. To keep the central k-space gap constant, for increasing TE, BW is reduced and thus encoding time increases. Acquisition parameters are listed in Table 1. Figure 1 conveys the FID for the healthy tooth specimen, where a rapidly decaying component with a T2* on the order of 10µs is observed. A dipolar oscillation is observed at 45µs, consistent with previous observations of collagen-rich specimens[11]. The slower decay of bound water dominates after the oscillation.
Figure 2A presents in vitro images of both tooth specimens for selected TEs as well as the difference image between TE = 10.9µs and TE = 25.0µs. All images show high signal intensity, but considerable T2-blurring is observed except for TE = 25.0µs. This effect stems from significant signal decay over the encoding time: first the ultra-fast decay of hard tissue at TE = 10.9µs and then the decay of bound water at TE = 105.4µs. The difference images show the ultra-short T2 components, but there is little contrast between structures such as enamel and dentin. The dental filling decays rapidly and is no longer observable by TE = 105.4µs, indicating that its chief component is the short-lived signal. The pulp cavity does not clearly exhibit longer-lived components and is thus hypothesized to have dried out in the storage process. Figure 2B represents the intensity acquired from ROIs drawn on enamel, dentin, and dental filling as a function of TE for the tooth specimen with filling. All regions present an ultra-fast decaying component, with a notably higher contribution in dentin. Dipolar oscillations are observed in both the dentin and enamel regions, with a higher prominence in dentin. Apart from tissue structure, these differences are likely affected by partial volume effects and varied T2-blurring behavior on the edges. Thereafter (>50µs), dentin and enamel decay apparently monoexponentially. The dental filling shows a slowly decaying component with a low signal contribution.
Figure 3 shows in vivo MRI of the teeth for two TEs as well as the accompanying difference image. Long-living, water- and fat-based signal components have been suppressed by the subtraction, leaving only the hard tissues in the teeth. This work presented the analysis of ultra-fast decaying MR signals in human teeth. Through both FIDs and multi-echo acquisitions with appropriate short-T2 hardware, hard tissues which decay on the order of 10µs were successfully identified and directly imaged. However, spatial resolution is limited by an encoding time still too long with respect to the signal decay. Resolution improvements are expected from further optimization on both the acquisition and processing side. Direct MRI of the hard tissues of teeth is possible and introduces avenues for numerous applications to explore.
Jason Daniel VAN SCHOOR (Zurich, Switzerland), Markus WEIGER, Emily Louise BAADSVIK, Klaas Paul PRÜSSMANN
14:05 - 14:15
#47026 - PG045 Frequency-dependent diffusion-relaxation correlation MRI: in vivo 2D multi-slice and ex vivo 3D sparsely-sampled acquisitions.
PG045 Frequency-dependent diffusion-relaxation correlation MRI: in vivo 2D multi-slice and ex vivo 3D sparsely-sampled acquisitions.
Diffusion MRI indirectly depicts the tissue’s microstructure at a few micrometers scale and characterizes it with quantitative metrics such as fractional anisotropy, mean diffusivity, or mean diffusion direction. However, its limited spatial resolution, coarser by 1 to 3 orders of magnitude than the diffusion extent, leads to voxel-averaged parameters and ambiguities in their interpretations. This length scale gap can be bridged using a diffusion tensor distribution (DTD) to describe the heterogeneity of the microstructure at a sub-voxel level[1]. Tensor-valued diffusion encoding eases the DTD determination by separating the isotropic and anisotropic contributions of the observed diffusivities[2]. Frequency-dependent diffusion encoding can also be added to the tensor-valued diffusion encoding to separate the restriction effects and account for the intrinsic frequency dispersion of the gradient waveforms[3,4]. The DTD can be correlated to the T1 and T2 relaxation times by using a variable TR and TE acquisition[5]. However, the concomitant sampling of all these parameters requires the acquisition of long series of one to several hundred contrasts, requiring single or few-shot EPI acquisitions to perform in vivo 2D or ex vivo 3D high-resolution imaging. Here, we intend to exemplify the possibilities offered by frequency-dependent diffusion-relaxation correlation to provide a specific description of the tissue microstructure of in vivo[6] and ex vivo brain at a sub-voxel level, allowing MRI to challenge histology. We also intend to present various strategies for multi-slices and sparsely sampled acquisitions of such a massively multidimensional dataset.
Frequency-dependent diffusion-relaxation correlation datasets were acquired in vivo on rat brains at 7 T and ex vivo on mouse and pig brains at 11.7 and 4.7 T, respectively. Fig. 1a presents the SE-EPI sequence customized with tensor-valued diffusion encoding and variable echo and repetition times. Gradient waveforms for different encoding anisotropies (bΔ = -0.5, 0, and 1) and increasing modulation orders 0, 1, and 2 associated with their diffusion spectra are presented in Fig. 1b. The use of 0 and 1st order waveforms allows designing an acquisition protocol of 389 images leading to an acquisition duration of 17 minutes shown in Fig 1c compatible with in vivo conditions. Even with a gradient strength of 760 mT/m, diffusion frequencies can reach 100 Hz for b-values up to 2.1 ms/µm2 with the use of 1st order modulated gradient waveforms. The 10 and 90 percentiles of the frequency range achieved in vivo correspond to 18 and 92 Hz and are presented in Fig. 1d. Fig. 2 illustrates the diversity and the high quality of the quantitative parameter maps acquired with the Fig. 1 protocol on an in vivo rat brain at 7 T. The acquisition time was 1 hour and 10 minutes due to the use of two segments and reversed blip acquisitions, allowing top-up correction of B0 inhomogeneity artefacts. Diffusion frequency dependence is especially prominent in the cerebellum and olfactory bulb gray matter.
Fig. 3 shows a slice of an ex vivo mouse brain acquired at 11.7 T in 3D at 150 µm isotropic resolution. To decrease acquisition time, the dataset was undersampled from 100 to 12.5 % retrospectively to test the efficiency and accuracy of a locally low rank reconstruction performed with BART. Fig. 4 compares various acceleration factors on 3D phase-encoded SE-EPI image series by using kY, kZ sampling along the echo train on a pig brain and regularization in the spatial and contrast dimensions. Frequency-dependent diffusion-relaxation correlation can be performed in vivo on rat brains and leads to a large diversity of parameter maps enhanced by the possibility to bin the solution space and discriminate intra-voxel component specific to white matter, gray matter, and cerebrospinal fluid, even in heterogeneous voxels[6].
Ex vivo, acquisition times of 10 to 20 hours are extended to get isotropic high-resolution phase-encoded 3D images. Still, the use of SE-EPI remains mandatory to acquire a series of a few hundred images required by frequency-dependent diffusion-relaxation correlation. A factor of 8 in acceleration is possible even with a single-channel volume coil. The quantitative parameter maps are well preserved at such acceleration, even if the decrease in SNR hampers the quality of the parameter maps with the lowest dynamic range, such as the Δω/2πE[Diso] map. Such high acceleration factors can be used for reducing the echo train length, increasing acquisition bandwidth, and thus decreasing T2* broadening and B0 inhomogeneity artefacts in sparse-sampled SE-EPI images. Frequency-dependent diffusion-relaxation correlation increases MRI specificity by quantifying and correlating parameters at a sub-voxel level, allowing MRI to compete with histology. We believe that fast acquisitions allowed by EPI sequences and sparse acquisitions can be the key to broadening its applications and usability.
Maxime YON (Rennes), Omar NARVAEZ, Pierre-Antoine ELIAT, Alejandra SIERRA, Daniel TOPGAARD
14:15 - 14:25
#47921 - PG046 Agentic MR sequence development - Leveraging LLMs with MR tools and tests for physics-informed sequence development.
PG046 Agentic MR sequence development - Leveraging LLMs with MR tools and tests for physics-informed sequence development.
Magnetic Resonance Imaging (MRI) requires precise timing of radiofrequency pulses and magnetic field gradients. Traditional sequence development demands both physics expertise and programming skills, creating bottlenecks in MRI research. While Large Language Models (LLMs) can generate code, MRI's physical and hardware constraints present unique challenges, as demonstrated in our previous work [1]. Small errors in timing, gradients, or RF pulses can cause contrast issues, artifacts, or signal loss, making simple code generation inadequate for reliable sequences. We present an agentic workflow combining LLMs with MR-specific tools and tests. Similar to test-driven development, this system enables physics-informed sequence generation and validation using the `pypulseq` library, iteratively refining sequences until they meet all requirements.
Our agentic system (Figure 1) combines LLMs with MR physics knowledge and `pypulseq` (v1.4.2) expertise. The system uses a feedback loop with four key validation tools:
1. Code Execution: Catches syntax errors and hardware violations
2. Timing Analysis: Verifies hardware raster times and sequence parameters
3. k-space Analysis: Validates trajectory coverage and gradient moments
4. Image Simulation: Detects subtle artifacts and contrast issues
The agent iteratively generates and refines code until meeting validation criteria, implemented in "Cursor AI". We evaluated the system using a standardized task: implementing a spin echo EPI sequence with 100ms TE. The goal was generating an artifact-free `.seq` file across different base LLMs.
To evaluate the agentic workflow, we tested the task
"Please, first read the LLM4MR instructions. Now, please code a spin echo EPI with a TE of 100 ms. Finish only when you are 100% sure it is properly implemented. Use the terminal to test the codes. Use all tools to validate the sequence."
We measured success by counting user interactions needed to achieve a satisfactory sequence. Each interaction represents a hint or instruction for code modification. The ideal case requires only the initial task prompt. While we must admit that this agent and the abstract was created very shortly before the ESMRMB deadline, we were quite surprised about the poor results of the agentic MR assistant, as shown by the results in Figure 2. Especially in the context of a much simpler setup tested recently (see https://www.mr-physik.med.fau.de/2025/03/03/mr-physicists-last-exam-llm4mr/), in which the LLMs were at least successful for some trials, without any additional tools.
We still think it is interesting to share with the community that the additional tools seem to have confused the LLMs, and often lead to endless thoughts about errors reported by our timing or k-space trajectory tests. Thus we conclude that such a setup requires further refinement, which we will test until October.
Coding errors were definitely solved always automatically, but sequence programming often got stuck when e.g. k-space sampling was off, see one intermediate outcome that it could never fix shown in Figure 3, even with all tools and tests. Our presented agentic workflow, integrating an LLM with `pypulseq` and a suite of MR-specific analysis and simulation tools, was thought to be a viable path towards robust, physics-informed sequence generation. The iterative loop, leveraging feedback from k-space analysis, timing checks, sequence reports, and crucially, MR simulation, was thought to allow the agent to identify and correct errors that would render a sequence unusable.
However, it turned out the tools helped eliminating coding errors, but otherwise lowered the performance of the pure LLMs strongly, which has still to be investigated in more detail.
Better explanation for the LLM how to use the tools and to interpret errors, or using MPC connection and multiple agents might improve the performance. We think agentic sequence development is the way to go and we have a promising toolbox and testbox, yet we could not yet get it to run properly and will be happy to report about our fails and progress at the meeting.
Moritz ZAISS (, Germany), Jonathan ENDRES, Simon WEINMÜLLER
14:25 - 14:35
#47738 - PG047 Is PCASL suitable for assessment of myocardial perfusion?
PG047 Is PCASL suitable for assessment of myocardial perfusion?
The evaluation of regional blood flow in the heart muscle is important for clinical diagnosis of cardiac diseases. Various arterial spin labeling (ASL) techniques (pulsed ASL, velocity-selective ASL) have been used for non-invasive quantitative evaluation of myocardial perfusion [1-3]. We recently demonstrated that pseudo-continuous arterial spin labeling (PCASL) provides high quality perfusion images of the lungs [4]. In this work, we present the first results of PCASL imaging to measure myocardial perfusion using different labeling strategies.
Measurements were performed on a 1.5 T MR scanner (MAGNETOM Aera, Siemens Healthineers AG, Forchheim) using a PCASL sequence with ECG-triggered labeling pulses and a bSSFP readout. The labeling plane was placed nearly perpendicular to the left ventricular output tract (LVOT) and the labeling pulse was triggered by the scanner’s ECG device (Figure 1). Different labeling durations (LDs, 300-1200 ms) were used and the bSSFP readout was performed in diastole by using suitable post labeling delays (PLDs, 300-600 ms). Images of the short cardiac axis and of the LVOT were acquired. Each measurement, consisting of 16 label/control image pairs and a proton density weighted image, was performed with a repetition delay of ≥ 5 s under synchronized breathing conditions. Depending on the PLD and cardiac cycle, the measurement time was around 3-4 min. PCASL images were registered non-rigidly using an optical flow-based image registration approach [5]. Other sequence parameters were: TR of 2.3 ms, TE of 0.9 ms, 70° flip angle, 8 mm slice thickness, 2.5×2.5 mm2 pixel size, 128×96 matrix, 930 Hz/pixel bandwidth, 160 ms acquisition duration. In Fig. 2, PCASL images of LVOT of a healthy subject are shown obtained with an LD of 300 ms and a PLD of 300 ms. The large difference in blood signal in the ascending aorta between control and labeling images indicates that the magnetization of the blood flowing through the LVOT is effectively inverted by the labeling pulse. A corresponding color-coded perfusion-weighted image is shown in Fig. 3 on the left. Due to the short LD and PLD, a “bolus” of labeled blood is generated in ascending aorta. In contrast, only a low perfusion signal (PWS) is observed in the myocardium, as the labeled blood has not yet been able to reach the heart muscle within a short PLD of 300 ms. With a longer LD of 1200 ms (covering two systoles), a relatively higher perfusion signal could be measured in myocardium, as more labeled blood reaches the heart muscle (Fig. 3, right). Fig. 4 shows a color-coded PCASL perfusion-weighted image of the heart short axis. A small but distinct difference between control and labeling signal values can be seen (Fig. 4, right). About 5% of the cardiac output flows through the heart muscle. However, complex blood flow pathways, high pulsatility of flow as well as respiratory and cardiac movements make ASL measurements of myocardial perfusion a very demanding task. Our preliminary results show that the ECG-triggered PCASL bSSFP approach can effectively label the blood flow in the ascending aorta. We further found that longer LD is required to improve the measurement of myocardial perfusion. PCASL bSSFP imaging of myocardial perfusion is feasible but further systematic measurements are needed to increase myocardial perfusion signal.
Petros MARTIROSIAN (Tübingen, Germany), Anja HANSER, Rolf POHMANN, Martin SCHWARTZ, Cecilia LIANG, Thomas KÜSTNER, Fritz SCHICK
14:35 - 14:45
#46517 - PG048 Proof of concept study for Deuterium Metabolic Imaging (DMI) in human breast.
PG048 Proof of concept study for Deuterium Metabolic Imaging (DMI) in human breast.
Breast cancer is the most common cancer in women and new therapeutic options require early and specific evaluation of treatment response. However, specific non-invasive imaging is still limited or technically demanding, preventing further implementation in the clinical domain. Deuterium metabolic imaging (DMI) is a new metabolic imaging technique where 2H MRSI is combined with the administration of 2H labeled substrates, that could potentially close this gap [1, 2]. In breast cancer, 2H MRS has been used to study [2H9]choline metabolism in an animal model and tumor cell death using [2,3-2H2]fumarate; but DMI has never been applied to the human breast to date to the best of our knowledge [3, 4].
Hardware: Experiments were performed at 7 T (Philips, Best, The Netherlands) using a double-tuned 2H/31P whole-body birdcage coil for 2H transmit integrated behind the bore of the MR system [5] and a single-tuned 2H loop coil for 2H receive tuned to 45.7 MHz. Two proton dipole antennas were used for transmit/receive for 1H MRI and B0/B1 shimming. Subjects were scanned in prone position (Fig. 1).
Phantom/healthy subjects: A flask containing mostly water and a small fat phase of rice oil in an inner sphere served as phantom. In vivo measurements were performed on two healthy volunteers (26 and 36 years), one was only scanned at 2H natural abundance (NA), one was scanned at NA and after drinking of 0.75 g/kg body weight [6,6-2H2]glucose (Glc; Buchem, Apeldoorn, The Netherlands) after an overnight fast whereby two 2H MRSI scans were started 60 and 81 min after drinking. Measurements were approved by the local ethics committee and written informed consent was obtained.
1H MRI and DMI acquisitions: At first, B1 and 2nd order B0 shimming were performed with volumes of interests covering a small central portion of the breast and the entire breast. Axial and coronal T1-weighted anatomical reference images were obtained using 2D multi-slice gradient echo sequences matching the field-of-view and the number of slices of the 2H MRSI scan. Axial 2-point Dixon-type sequences were obtained. 2H MRSI were acquired using a 3D FID-MRSI sequence employing Hamming-weighted k-space sampling (TR/TE 100/1.37 ms, nominal flip angle 40°, NSA 20, FOV RLxAPxFH 420x220x340 mm³, nominal voxel size 20x20x20 mm³, spectral bandwidth 2750 Hz, spectral points 256, scan duration 21:01 min). 2H MRSI data were reconstructed by Fourier-transformation in the spatial and spectral domains with in-house written scripts in MATLAB R2020b (TheMathWorks, Natick, MA, USA). Phantom measurements demonstrated the detectability of 2H water signal from NA (HDO), set to 4.7 ppm. In voxels containing water and rice oil, also small lipid signals (1.0-1.3 ppm) could be detected. In vivo 3D 2H MRSI measurements in healthy volunteers of the right breast showed different HDO/lipid signal ratios, depending on the voxel composition of fat and glandular breast tissue (Fig. 2). After oral administration of [6,6-2H2]Glc, a new signal (upfield shoulder peak) was detected in the right breast after 60 min at 3.8 ppm, corresponding to an overlapping Glc signal (Fig. 3). In Fig. 4 A/B, the 2H spectra of a breast voxel before and 60 min after Glc administration were superimposed and the difference spectrum calculated, with evidence of the resulting Glc signal. The increase in the lipid signal is most likely due to subtraction artifacts, since the Glc was administered after the NA scan outside the scanner. Fig. 4 C/D shows 2H spectra of the same voxel 60 and 81 min after Glc showing a further increase of Glc, an increase in HDO, and no signal detectable at the lipid position. As previously in DMI studies in the brain or liver [1, 5], we were able to detect an HDO signal of 2H NA also in the breast. Furthermore, after oral ingestion of Glc, a corresponding Glc signal was detectable, overlapping with the HDO signal, demonstrating detectable Glc metabolism, consistent with results from 18FDG-PET [6]. Based on the SNR of the Glc in the breast, the pectoral or heart muscle and the relative spatial distances between them, the Glc breast signal cannot be explained by overlapping point-spread functions of signals from those tissues associated with low resolution MRSI. In contrast to previous DMI applications, in the breast, the 2H NA lipid signal is detectable in the organ of interest and not only in adjacent tissues. Due to the location of the lipid signal, this will particularly complicate the detection of lactate. However, without repositioning between the two 2H MRSI scans after Glc administration, we were unable to detect any subtraction artifact of the lipid signal. Thus, lactate detection could be feasible. A possible glutamate/glutamine Glx signal presumably overlaps with the left shoulder of the lipid signal. Our results demonstrate the technical feasibility of DMI in the human breast as a starting point for initial measurements in patients with cancer, but requires further validation.
Claudius S. MATHY (Erlangen, Germany), Luka STAM, Mark GOSSELINK, Sonja VLIEK, Dimitri WELTING, Cezar ALBORAHAL, Michael UDER, Tobias BÄUERLE, Armin M. NAGEL, Jannie P. WIJNEN, Dennis W.j. KLOMP
|
Auditorium 900 |
|
"Saturday 11 October"
Added to your list of favorites
Deleted from your list of favorites
C34
13:45 - 14:45
FT2 Oral - Imaging advances
For clinical solutions throughout the body
ET2: Cycle of Clinical Practice
13:45 - 13:55
#45789 - PG049 Investigating the microscopic diffusion MRI properties of prostate tissue using MR microscopy.
PG049 Investigating the microscopic diffusion MRI properties of prostate tissue using MR microscopy.
Diffusion MRI (dMRI) plays a key role in the diagnosis of prostate cancer[1]. Current research is focused on quantitative dMRI techniques that use mathematical signal models to measure the diffusion properties of prostate tissue[2]. The measurements obtained through these techniques could act as quantitative biomarkers for prostate cancer. However, the relationship between the diffusion properties and histological features of prostate tissue is currently speculative and requires empirical validation.
The key components of prostate tissue are epithelium, stroma, and lumen (ESL). Prostate cancer is associated with the proliferation of epithelial cells, with invasive growth into luminal space and stroma[3]. An improved understanding of the diffusion properties of each tissue component could help to improve the specificity of quantitative dMRI measurements to cancer.
Previous studies have used optical microscopy of tissue samples to correlate dMRI measures with histological features[4–6]. However, this approach is constrained to two-dimensional analysis and is limited by image registration challenges[7]. Microscopic resolution MRI (MR microscopy) could alleviate these challenges, as it enables the structures of a prostate tissue sample to be resolved in 3D, in exact spatial alignment with the diffusion images used for modelling[8–10].
We aim to use MR microscopy to distinguish the ESL components of prostate tissue samples and study the diffusion properties of each component.
Nine samples of prostate tissue (~3mm diam. 4mm length) were obtained from prostatectomy surgery of two patients. Samples were fixed in formalin and equilibrated in 0.5ml/L Gadolinium solution.
MR imaging was performed using a 16.4T Bruker system. Table 1 displays the sequence parameters for the images acquired of each sample.
The DTI and MGE images (20µm and 40µm resolution, respectively) were used for ESL segmentation. Fractional anisotropy (FA) and mean diffusivity (D) maps were calculated for each sample. Regions with high DxFA were classified as stroma[9]. Lumen was classified as regions with MGE signal intensity (avg. over TE) similar to the surrounding medium. All other regions were classified as epithelium, provided the MGE signal intensity was above a minimum threshold. Figure 1 displays example ESL segmentations for two samples.
Multi-b-value and multi-∆ dMRI images were acquired at lower resolution (480x480x320µm). For each image, the normalized signal intensity of each voxel was represented as a weighted linear sum of the ESL volume fractions calculated using the high-resolution segmentations. This linear model was optimized for all voxels within the samples; the optimized weights represent the average dMRI signal intensities for the ESL components. This was repeated for all images to obtain ESL dMRI signal measurements at varying b-value and ∆. Uncertainties on each signal measurement were estimated through bootstrapping.
Five isotropic models were fit to the signal measurements for the ESL components using a least-squares fitting method (Table 2). Uncertainties in signal measurements were propagated into model parameter uncertainty using the Jacobian of the model fitting function. The Akaike information criterion (AIC) was calculated to compare the relative information content of models. Epithelial (E) signals were found to be higher than stromal (S) signals (Figure 2), consistent with previous work[9]. For both E and S, higher signals were measured for sequences with long ∆, however this difference is larger for E. The sphere + ball models (4 and 5 param) provide the best fit to the measured signals (lowest AIC values). Within the 4 parameter model, the sphere radius estimates are similar for E and S (approx. 7µm), but a higher sphere volume fraction and lower diffusivity are observed for E (Table 2). This project presents a novel method for separating dMRI signal contributions from the microscopic components of prostate tissue at varying b-value and ∆. The raised signals measured at longer ∆ reflect physical restriction of water diffusion within the tissue. Models including the ‘sphere’ function[11] depend on ∆ so can describe the measured signals better than the ADC and DKI models.
These results could help to refine and regularize models used for in vivo quantitative dMRI (e.g. VERDICT[12], RSI[13], and HM-MRI[14]) and strengthen the link between dMRI measurements and the biological changes associated with cancer; however, the effects of tissue fixation (diffusivity reduction and cell shrinkage) need to be considered. Histological processing of the samples is in progress; this will enable the dMRI properties of E and S at different Gleason grades to be assessed. Further prostatectomy surgeries are scheduled in the coming months, from which more tissue samples will be collected and imaged. Measured dMRI signals for epithelium and stroma indicate physical restriction of water diffusion and are best described by sphere + ball models.
Adam PHIPPS (London, United Kingdom), Nyoman KURNIAWAN, David ATKINSON, Panagiotaki ELEFTHERIA, Roger BOURNE
13:55 - 14:05
#46940 - PG050 A fast MRI-based HIFU beam refocusing method for optimized acoustic energy deposition in several locations.
PG050 A fast MRI-based HIFU beam refocusing method for optimized acoustic energy deposition in several locations.
MRI-guided focused ultrasound (HIFU) is a non-invasive therapy. However, the speed of ultrasound propagation varies depending on the biological tissue (skin, muscle, fat, etc.). This can lead to suboptimal focusing at the target site. The aim of this work is to present an MRI-based calibration method for the correction of these aberrations. Displacement maps calculated from fast MR-ARFI images allow the calculation of amplitude and phase corrections to be applied to each transducer element of the HIFU device. These corrections are used to improve the focusing quality.
The MRI-ARFI sequence [1] is a single-shot echo planar imaging (68x64 matrix, 2.5x2.5x3 mm³ resolution, TE/TR=27/1500 ms, 70° FA, partial Fourier 6/7, 1.5T Avanto fit, Siemens Healthineers, Germany). It integrates a bipolar motion-encoding gradient (5 ms per lobe), the first lobe of which is synchronised with a 5 ms ultrasound pulse. The HIFU device consists of a 32-element transducer (f/D=13/13 cm, 1 MHz operating frequency, Imasonic, France). It is driven by a programmable generator (Image Guided Therapy SA, France). The amplitude and phase are adjustable for each channel. The experiments were carried out on an ex vivo muscle of the sheep. The sample was immersed in degassed water and positioned in front of the transducer.
The calibration experiment was based on the Hadamard coding technique [2], in which 128 ultrasound pulses are applied and the resulting tissue displacements are imaged by means of fast MRI ARFI. An inverse method is then used to calculate the amplitude and phase corrections to be applied to each transducer element in order to refocus the HIFU beam at a desired location. This is shown in Figure 1. For this purpose, the displacement values measured in a single pixel for each ultrasound condition during Hadamard encoding are plotted and serve as input values for the calculation of the inverse solution for the calculation of the amplitude and phase corrections. A comparison was made of the displacement values at the target pixel during HIFU sonication, before and after correction. Figure 2A shows the displacement map for the initial condition (no correction, focal spot positioned 3 cm beyond the imaging slice). From this image, three pixels intercepting the ultrasound beam and separated by 7.5 mm each were selected to refocus the beam at each location from a single Hadamard calibration experiment. HIFU focusing was systematically improved after phase correction (B, C, D), showing a smaller displaced spot compared to the initial experiment (A). The gain in displacement with respect to power was about 50% in all cases (F, G, H vs. E) when only phase corrections were applied, and reached 60% when both amplitude and phase corrections were applied. The proposed calibration method allows the optimisation of the acoustic energy delivery at the desired location in combination with a fast MR-ARFI sequence in a clinical MRI scanner. By shortening the sequence repetition time (here 1.5 s to maximise SNR and evaluate the refocusing method), the total acquisition time of the calibration experiment (here 9 min 30 s) can be further reduced. The targeting of the focal spot beyond the position of the MR image slice during Hadamard encoding allows the calculation of different corrections in amplitude and phase to improve the focus at multiple locations (here over a distance of 1.5 cm, see Figure 2) from a single calibration experiment. The proposed method may be valuable for improving the efficiency of MR-HIFU therapies by increasing the local acoustic energy deposition at the desired location. MR-ARFI in combination with Hadamard coding provides a non-invasive method to compute amplitude and phase corrections to be applied to each HIFU transducer element to compensate for propagation aberrations in a biological tissue. Since current clinical applications of MR-HIFU often require treatment of pathological regions larger than the focal spot dimensions, the ability to provide corrections at multiple locations from a single calibration experiment is of key interest. Furthermore, the delivery of excessive acoustic energy in the near-field of the HIFU beam, which can cause unwanted side effects such as skin burns, is avoided by optimising the quality of the focus.
Fanny DABRIN (Bordeaux)
14:05 - 14:15
#46031 - PG051 Intraepineurial fat fraction: A novel MR Neurography-based biomarker in Transthyretin amyloidosis polyneuropathy.
PG051 Intraepineurial fat fraction: A novel MR Neurography-based biomarker in Transthyretin amyloidosis polyneuropathy.
Hereditary transthyretin amyloid polyneuropathy (ATTRv-PN) is a rare, progressive axonal neuropathy, for which early detection is critical. However, standard assessments, including nerve conduction studies, often lack sensitivity for subclinical disease [1].
Quantitative MRI (qMRI) has emerged as a tool to monitor nerve and muscle changes in neuromuscular disorders [2,3], using metrics like fat fraction (FF), volume, and magnetization transfer ratio (MTR). While FF is widely used in muscle imaging, its application to peripheral nerves is rare [4].
Recent histological and biochemical studies revealed lipid droplets within amyloid deposits, especially in the endoneurium, and a key role for lipid–amyloid interactions in fibril aggregation [5,6].
These findings support the hypothesis that intraepineurial fat fraction (ieFF), measured by MR neurography, may reflect both amyloid-related lipid deposition and nerve fiber loss with epineurial remodeling, making it a promising imaging biomarker in ATTRv-PN.
In this study, we aimed to determine if ieFF is a relevant and sensitive marker of early nerve damage, compare it to MTR and nerve volume, and evaluate its association with clinical and electrophysiological severity, assessing ieFF in sciatic and tibial nerves using qMRI in ATTRv-PN patients, asymptomatic carriers (ATTRv-C), and healthy controls (HC).
53 TTR mutation carriers (31 ATTRv-PN, 22 ATTRv-C) and 24 controls underwent lower-limb qMRI. Sciatic and tibial nerves were manually segmented (Figure 1), and Dixon based-ieFF, MTR, and volume were extracted.
Clinical scores included ONLS, RODS, NIS-LL, MRC; electrophysiology included CMAP, MUNIX, SNAP.
Univariate and multivariate models (adjusted for age and BMI) were used to assess the diagnostic and prognostic value of ieFF. ieFF was significantly increased in ATTRv-PN nerves vs controls: sciatic (32.4% vs 22.3%) and tibial (13.7% vs 9.74%, p < 0.01). ATTRv-C also showed elevated ieFF (p < 0.05), despite normal clinical scores (Figure 2).
In multivariate correlation model, ieFF was the only imaging marker independently associated with ONLS, RODS, MRC, and CMAP (Figure 3). In contrast, MTR and volume had no significant correlations with severity.
ieFF also correlated with volume and inversely with MTR, reinforcing its biological plausibility.
In ATTRv-PN, there was reduced tibial MTR and increased nerve volume, but these changes were not associated with severity. ATTRv-C displayed an intermediate profile: ieFF values close to ATTRv-PN, but normal MTR and volume, distinguishing them from both patients and controls.These findings highlight ieFF as a robust biomarker for diagnosis and monitoring, even in presymptomatic stages. Intraepineurial fat fraction (ieFF) is a novel and sensitive qMRI biomarker in ATTRv neuropathy. It distinguishes symptomatic patients and asymptomatic carriers from healthy controls and correlates robustly with clinical severity. Unlike MTR and volume imaging markers, ieFF retained independent associations in multivariate analysis and appears to reflect early nerve involvement. Its histological plausibility, non-invasive acquisition, and strong correlations with functional impairment highlight its potential for clinical research. Importantly, ieFF reveals an intermediate imaging profile in ATTRv-C, not captured by MTR or volume metrics, supporting its value in characterizing subclinical stages of the disease. The present results illustrate the interest of a multimodal qMRI imaging analysis of the whole lower limb in patients with ATTRv polyneuropathy while highlighting ieFF as a new sensitive biomarker. This parameter is able to distinguish ATTRv-PN and ATTRv-C patients from healthy controls and was highly correlated with the main electrophysiological and clinical severity scores. This nerve fat infiltration would likely illustrate the lipid droplets found in endoneurial amyloid deposits and the onset of axonal loss accompanied by a high-fat epineurial connective tissue replacement at a sub-clinical level.
Eva SOLE CRUZ (Marseille), Etienne FORTANIER, Constance P. MICHEL, Emilien DELMONT, Annie VERSCHUEREN, Marc-Adrien HOSTIN, David BENDAHAN
14:15 - 14:25
#47403 - PG052 Multiparametric quantitative MRI with oxygen extraction fraction: Prospective characterisation of soft tissue sarcomas, prediction of radiotherapy response and correlation with multiplex immunofluorescence.
PG052 Multiparametric quantitative MRI with oxygen extraction fraction: Prospective characterisation of soft tissue sarcomas, prediction of radiotherapy response and correlation with multiplex immunofluorescence.
Soft tissue sarcomas (STS) are rare heterogeneous tumors, with few predictive factors of response to neoadjuvant radiotherapy (nRT) despite important variations [1]. Hypoxia is a predictive and prognostic factor in many cancers, but its evaluation by nitroimidazole PET CT remains poorly available. Our interventional trial (NCT05684874) aims to evaluate the feasibility, characterization interest and predictive value of a multiparametric quantitative MRI (mpqMRI) including quantification of relative oxygen extraction fraction (rOEF) without contrast agent injection.
Adult limb and trunk STS patients were prospectively included and performed mpqMRI pre- and post-nRT. Acquisitions were performed on a Siemens MAGNETOM Vida 3T clinical MRI (Siemens Healthineers, Erlangen, Germany). The research protocol duration was 14 min and included 3 sequences. The first one was a chemical-shift encoded 3-dimensional fast low-angle shot multi-echoes gradient-echo sequence (3D FLASH CSE-MRI; n=10 echoes), with an echo spacing of 1.2 ms to properly separate fat and water [2]. It allowed measurement of R2*, magnetic susceptibility and proton density fat fraction (PDFF). R2* was calculated after correction of chemical-shift and B0 macroscopic inhomogeneity. Then a 2-dimensional turbo spin echo multi-echo (TSE) was acquired with 10 echoes ranging from 8 to 80 ms. To avoid R2 underestimation, the first echo was discarded before performing a mono-exponential fitting. The last sequence was a multi-b-values diffusion-weighted pulse sequence (n=10 b-values, from 0 to 800 s.mm-2), allowing Bayesian inference of IVIM parameters. Relative blood volume (rBV) was approximated by the perfusion fraction f of the IVIM model, corrected by T2 and normalized by water fraction (1-PDFF). To ensure consistent data acquisition and avoid registration and resampling uncertainties, the research sequences were acquired with identical geometry. rOEF and SvO2 (calculated by substracting rOEF to the pre-MRI digital SpO2) were evaluated by quantitative blood oxygen level dependant method (qBOLD) using an adaptation of the model proposed by Toth et al. [3]:
rOEF=SaO2-SvO2= (R2*-R2)/(4/3·π·γ·Δχ0.Hct·B0.rBV)
With Δχ0 the difference of magnetic susceptibilities between fully oxygenated and fully deoxygenated haemoglobin, B0 the magnetic field strength, γ the gyromagnetic ratio of the proton. The small vessel hematocrit Hct was approximated as 75% of the macrovascular hematocrit measured on a pre-MRI total blood count.
Median value of each parameter in the volume of interest was recorded, and the correlation between these values and the percentage of viable cells on the surgical specimen (a prognostic factor) was calculated. Furthermore, 2 areas of the surgical specimen were identified for analyses in multiplex immunofluorescence (mIF), with a panel including, among others, CA9 and CD34 antibodies to stain hypoxic cells and endothelial cells, respectively. Fifteen STS patients were included, and 3 of them were secondarily excluded due to surgical refusal (n=2) or metastatic progression (n=1). The mpqMRI protocol could be performed in every patient and without significant artefacts (Figure 1). Overall, the tumors were not hypoxic, with pretreatment median rOEF always <22%. This result was confirmed by mIF, with a single sample presenting a density of CA9+ cells >3/mm2 (Figure 2). After nRT, significant decrease of T2* and SvO2 and increase of R2’, ADC and Dslow were observed (Figure 3). The percentage of persisting viable cells tended to be predicted by the pretreatment median SvO2 (Rho=-0.53, p=0.08; Figure 4), and was correlated with post-nRT R2’, rOEF and SvO2. These STS mpqMRI measures are the first available in literature but are consistent with usual biological data, reported in prostate or muscles. Measure of mpqMRI parameters modification during cancer treatment may help predict the treatment response and adapt patient management.
This study has limitations. These preliminary results are reported from the median values of whole tumor, a regionalized analysis from each quantitative map will improve the constancy by accounting for tumor heterogeneity in the analysis. In addition, accuracy, reproducibility and repeatability are ongoing using Calimetrix® PDFF-R2* phantom, CaliberMRI® Diffusion phantom, and a T2 phantom made by Centre de Résonance Magnétique des Systèmes Biologiques (Bordeaux, France). Potential confounding factors that could influence R2’ and haemoglobin dissociation curve were not considered. An mpqMRI protocole evaluating rOEF is feasible in limb or trunk STS in a clinically acceptable time. Some quantitative features being potentially predicitive of treatment response. These promising results could allow treatment personalization trials and imaging biomarker developments.
Benoît ALLIGNET (Lyon), Benjamin LEPORQ, Floriane IZARN, Amine BOUHAMAMA, Frank PILLEUL, Alexandra MEURGEY, Gualter VAZ, Alexandre BEIGE, Marie-Pierre SUNYACH, Waisse WAISSI, Olivier BEUF
14:25 - 14:35
#47563 - PG053 Reinforcing the generalizability of spinal cord multiple sclerosis lesion segmentation models.
PG053 Reinforcing the generalizability of spinal cord multiple sclerosis lesion segmentation models.
Spinal cord (SC) imaging has become increasingly central in the diagnosis and monitoring of multiple sclerosis (MS) [1,2]. SC lesions bear strong prognostic significance, with evidence linking their spatial distribution to clinical disability [3–5]. Accurate segmentation of SC lesions is essential for monitoring disease progression. Moreover, despite recent initiatives [6–9], there remains a wide variability in MRI acquisition parameters across institutions. Existing SC lesion segmentation methods lack accessibility [10–12], are typically contrast-specific and often fail to generalise to previously unseen imaging protocols [13–16]. Additionally, inter- and intra-rater variability hinders the precise tracking of lesion changes. Our objective is to develop a robust model for MS lesion segmentation on MRI scans that generalises across different contrasts and imaging parameters. We explore two methods to improve generalizability compared to the current state-of-the-art methods.
A multi-site dataset (20 sites, 1850 people with MS, 4430 scans) was selected based on the heterogeneity in acquisition parameters and sequences: T1w spin echo (n=23), T2w (n=3061), T2*w (n=548), PSIR (n=363), STIR (n=92), MP2RAGE-UNIT1 (n=343) acquired at 1.5T and 3T on GE, Siemens and Philips MRI systems. The field-of-view coverage varied across sites (brain and upper SC, or SC only), and acquisitions were either 2D (axial: n=2895, sagittal: n=1169) or 3D (n=366), with voxel dimensions ranging from 0.2x0.2x5 mm3 to 0.8x0.8x9 mm3. Manual segmentations were collected from expert raters across multiple institutions. We explore the following strategies: (i) Weighted batch sampling: In each training batch, images are sampled with probabilities inversely proportional to the square root of the number of samples in each contrast, thereby up-weighting under-represented contrasts [17] ; (ii) Pretrained model fine-tuning: We fine-tuned a foundational model, pretrained on over 10,000 CT scans [18], on our multi-contrast dataset. Models were trained under equivalent hyperparameters for fair comparison. Evaluation employed both voxel-wise metrics (Dice coefficient) and lesion-wise metrics (lesion-wise positive predictive value (L-PPV), sensitivity, and F1-score). The results were benchmarked against existing SC lesion segmentation tools available in SpinalCordToolbox (SCT): (a) sct_deepseg_lesion for T2w/T2*w [13], (b) sct_deepseg for PSIR/STIR [16], and (c) sct_deepseg for MP2RAGE-UNIT1 [19]. Both experiments performed better than the baseline model. The average Dice score increased from 0.42 (baseline) to 0.44 with weighted batch sampling (i), and to 0.50 with CT-pretrained model fine-tuning (ii). Fine-tuning yielded the highest performance across most metrics, including Dice, L-PPV and L-F1. Interestingly, weighted sampling yielded slightly higher lesion sensitivity, indicating a trade-off between precision and recall. Contrast-specific analyses revealed strong improvements on under-represented modalities. For PSIR (8% of the dataset), Dice increased from 30.6% (baseline) to 45.8% (ii); for STIR (2% of the dataset), from 27.9% to 59.4% (ii). Even high-frequency contrasts (T2w and T2*w) showed performance gains (+8.8% and +1.3%, respectively) with (ii). Compared to state-of-the-art models, (ii) outperformed (a) and (c) on their respective contrasts. However, it did not surpass (b), which had partial access to the test data during training. Both weighted batch sampling and pretrained model fine-tuning independently improved generalisation, particularly benefiting under-represented contrasts. Our evaluation remains limited as methods (a), (b) and (c) were trained on some of the data used during testing, limiting fair comparisons. Moreover, Dice score, while widely used, is suboptimal for small lesions with uncertain boundaries [20]. Although lesion-wise metrics (L-PPV, L-F1) provide more lesion-centric insight, they rely on binary overlap thresholds and are susceptible to segmentation variability. In [21], we demonstrated that expert neuro-radiologist ratings, using a 1-5 Likert scale, often contradicted voxel-wise metrics: predicted segmentations were sometimes judged to better represent lesion presence than manual annotations, reflecting rater variability [11]. This highlights the need for complementary evaluation frameworks. In [21], we suggested that soft segmentations can improve clinical interpretability and enhance lesion detectability [21]. Nonetheless, expert review remains resource-intensive, emphasising the necessity of developing scalable surrogate evaluation metrics that better correlate with expert review. Fine-tuning a pretrained CT-based model yielded the best segmentation performance across diverse MRI contrasts, demonstrating the feasibility of cross-modality transfer learning in SC MS lesion segmentation. The model and code will be released as part of SCT, promoting reproducibility and collaborative development.
Pierre-Louis BENVENISTE (Montréal, Canada), Lisa Eunyoung LEE, Alexandre PRAT, Zachary VAVASOUR, Roger TAM, Anthony TRABOULSEE, Shannon KOLIND, Jiwon OH, Michelle CHEN, Charidimos TSAGKAS, Christina GRANZIERA, Nilser LAINES MEDINA, Mark MUHLAU, Jan KIRSCHKE, Julian MCGINNIS, Daniel S. REICH, Christopher HEMOND, Virginie CALLOT, Sarah DEMORTIÈRE, Bertrand AUDOIN, Govind NAIR, Massimo FILIPPI, Paola VALSASINA, Maria A. ROCCA, Olga CICCARELLI, Marios YIANNAKAS, Tobias GRANBERG, Russell OUELLETTE, Shahamat TAUHID, Rohit BAKSHI, Caterina MAINERO, Constantina A. TREABA, Anne KERBRAT, Elise BANNIER, Gilles EDAN, Pierre LABAUGE, Kristin P. O'GRADY, Seth A. SMITH, Timothy M. SHEPHERD, Erik CHARLSON, Jean-Christophe BRISSET, Jason TALBOTT, Yaou LIU, Hervé LOMBAERT, Julien COHEN-ADAD
14:35 - 14:45
#47376 - PG054 Gadolinium-based contrast agents in the rat kidney: Insights from spatially resolved mass spectrometry and MRI.
PG054 Gadolinium-based contrast agents in the rat kidney: Insights from spatially resolved mass spectrometry and MRI.
Gadolinium-based contrast agents (GBCA) are routinely administered to enhance magnetic resonance imaging (MRI) and are generally considered among the safest classes of drugs. However, in the last decade, safety concerns have emerged due to prolonged presence of Gadolinium (Gd) in various organs, particularly in association with linear GBCA. [1] As primary organ of excretion, the kidneys generally showed more Gd presence than other organs [2, 3] but the exact location and speciation of renal Gd remains underinvestigated. This study aimed to map and quantify the spatial distribution of GBCA residues in rat kidneys following high-dose administration over the course of their elimination.
A total of seventy rats received one of six marketed GBCA (three macrocyclic GBCA [mGBCA], three linear GBCA [lGBCA]; cumulative dose: 4.8 mmol Gd/kg body weight) or saline (control). Rats were sacrificed at 5 days or 14 weeks (n=5 per group and time point). Right kidneys were snap-frozen in their native state, followed by analysis of Gd distribution and concentration using laser ablation inductively coupled plasma mass spectrometry (LA-ICP-MS) imaging and semi-automated multi-layer concentric sub-segmentation. Distributions of intact GBCA chelates were analyzed by matrix-assisted laser desorption ionization mass spectrometry imaging (MALDI-MSI). Left kidneys were fixed in 4% formaldehyde solution for ex vivo MR microscopy. T1 maps were acquired with a 2-channel volumetric transceiver RF coil on a 9.4T animal MR scanner (PharmaScan, Bruker BioSpin) by use of a rapid acquisition with relaxation enhancement (RARE) technique. Quantitative visualization and segmentation were performed using in-house developed software. At day 5, LA-ICP-MS showed Gd predominantly localized in the renal cortex (Fig. 1). Depth profiles revealed peak concentrations of 2538±1163 nmol Gd/g for mGBCA and 1862±939 nmol Gd/g for lGBCA in the cortex, with 10- to 100-fold lower concentrations in the outer and inner medulla (Fig. 2). No significant difference was observed in the distribution pattern or concentration range of Gd between the classes of mGBCA and lGBCA. Results from MALDI-MSI largely agreed with those from LA-ICP-MS imaging, which supports that the species of the present Gd was intact GBCA chelate (Fig. 3). Parametric T1 mapping revealed substantial T1 shortening in ex vivo kidneys at day 5 following GBCA compared with controls, which was most pronounced in the renal cortex (Fig. 4). The T1 shortening obtained for lGBCA (ΔT1=-39%) was stronger than for mGBCA (ΔT1=-11%). Moreover, T1 shortening was also observed in the surrounding formaldehyde solution of GBCA kidneys but not of control kidneys. On one hand, this aligns with results from MALDI-MSI, indicating that Gd resides in form of the water-soluble intact chelate, which was washed out from the tissue into the supernatant during fixation and storage. The stronger T1 shortening in formaldehyde surrounding mGBCA kidneys suggest a more thorough washout compared with lGBCA kidneys. On the other hand, the washout of GBCA prevented any conclusions from being drawn regarding Gd content within the renal tissue by means of MRI.
At week 14, LA-ICP-MS showed remaining Gd concentrations orders of magnitude lower in both cortex and medulla with greater elimination of Gd in the cortex - particularly in mGBCA kidneys (Fig. 1). Peak Gd concentrations in the cortex were 11±15 nmol Gd/g following mGBCA and 128±101 nmol Gd/g following lGBCA. In the medulla, peak concentrations were 16±11 nmol Gd/g following mGBCA and 17±11 nmol Gd/g following lGBCA (Fig. 2). Gd concentrations following mGBCA were significantly lower than following lGBCA. Notably, Gd distribution images showed relatively high concentrations in medullary areas following several GBCA (Fig. 1). In line with this, depth profiles confirmed relatively high concentrations in the region corresponding to the boundary area between the inner and outer medulla (Fig. 2). MALDI-MSI indicated residual Gd from mGBCA as intact chelate but remained inconclusive for lGBCA (Fig. 3). GBCA residues in the kidney exhibited a non-homogeneous distribution pattern, which changed during continuous elimination over 14 weeks. Macrocyclic GBCA showed more thorough elimination resulting in in significantly decreased Gd presence compared with linear GBCA, likely due to the high-stability chelates, as shown by MALDI-MSI. Ex vivo T1 MR microscopy demonstrated the potential to aid research on contrast agent safety, but it requires dedicated sample preparation approaches to preserve analyte biodistribution. Despite their presence in the kidney observed in our study, no indications of renal impairment following clinical application of GBCA have been reported to date.
Luis HUMMEL (Berlin, Germany), Janina BOYKEN, Axel TREU, Hubertus PIETSCH, Thomas GLADYTZ, Ehsan TASBIHI, Thoralf NIENDORF, Erdmann SEELIGER
|
Salle Major |
|
"Saturday 11 October"
Added to your list of favorites
Deleted from your list of favorites
E34
13:45 - 14:45
MS4 - Innovation across scales in MRI RF coil engineering
From basic research to market transformation
|
Salle 76 |
14:45 |
TIME FOR A BREAK
- Coffee and refreshments will be available at the cash bar.
|
15:00 |
"Saturday 11 October"
Added to your list of favorites
Deleted from your list of favorites
E35
15:00 - 16:00
MS7 - Multiorgan Quantitative T1 Mapping
In Systemic Inflammation
|
Salle 76 |
|
"Saturday 11 October"
Added to your list of favorites
Deleted from your list of favorites
A35
15:00 - 16:00
LT - Registered Reports and Project Abstracts
|
Auditorium 900 |
|
"Saturday 11 October"
Added to your list of favorites
Deleted from your list of favorites
C35
15:00 - 16:00
FT3 Oral - Optimizing the MR Signal
15:00 - 15:10
#47867 - PG055 Breast Digital Twin: simulating with MR-zero.
PG055 Breast Digital Twin: simulating with MR-zero.
MR-zero is an MRI simulation framework based on Phase Distribution Graphs (PDG) [1]. In recent years, it has proved itself a very useful tool by providing realistic previews and optimizations of many sequences with phantom or even in-vivo digital twin setups [2,3]. Most proofs of concept for this simulation focused on brain MRI. With this work we propose to use MR-zero to simulate for the first time breast MRI, while also experimenting with the newest MR-zero extension: simulation of off-resonance RF pulses [4]. The proposed breast digital twin phantom allows to simulate several artefacts usually seen in breast MRI, such as chemical shift artefacts, fat-water interference, asymmetric B0 shimming and areas of high signal intensity near the breast surface due to the proximity of the receiving coil [5].
In this work, the Phase Distribution Graph Simulation within the MR-zero framework was used for simulation [1,4]. For simulation, an in-silico breast phantom or breast digital twin is required. To create this, we used a coarse segmentation of fat, fibroglandular tissue (FGT) and muscle based on a frequency selective 3D CEST MRI scan and the method presented in [6]. Using segmentation masks, we assigned to each voxel literature values for T1, T2, frequency shift and diffusion coefficient [8-12]. For the phantom voxels that contained fat, the corresponding voxels in the B0 map where set with the frequency shift of the main fat peak at 7 T (-3.4 ppm -> −1013 Hz). Additionally, the phantom was altered to reproduce (i) realistic B0 and B1+ field inhomogeneity using the corresponding B0 and B1+ maps measured in the subject at 7T, (ii) B1- coil sensitivities with stronger sensitivity in the left breast periphery, emulating closeness of the breast to the receiving element.
In this work, a FLASH sequence coded with PyPulseq [7] is used with default parameters TE = 1.9 ms, TR = 8.16 ms, FOV = 350 mm x 350 mm x 4 mm, matrix = 128 x 128, flip angle = 8°, centric reordering and bandwidth = 800 Hz/pixel.
This sequence was simulated in a first exercise in three flavours: (i) FLASH, (ii) fat-sat prepared FLASH, (iii) FLASH using binomial water excitation pulses. The setup of these preparations is the same as used in [4].
Then, we simulate the FLASH sequence with 4 multiplicative bandwidth factors to visualize the fat voxel shift in the readout direction.
Finally, we modify the phantom to have voxels with overlapping fat and FGT. This phantom is used to simulate FLASH sequences with different echo times, with which we image with in-phase and opposed phase fat and water spins. In Fig. 1, the parameter maps for the phantom used in simulations are shown. In the T1 and T2* maps, 3 different structures can be identified: fat, fibroglandular tissue and muscle tissue, segmented using the method in [6].
In Fig. 2(a-c), we see the simulation of the FLASH sequence for the phantom set up with homogeneous ∆B0, B1 and coil sensitivity. The images presented are the result of simulating a simple FLASH excitation and readout, the same sequence with fat saturation and, finally, with water excitation. In 2a, all tissues emit high signal, the higher signal in FGT being due to its higher apparent T2. In 2b and 2c, the absence of fat signal and the brightness of FGT, respectively, reveal that fat saturation and water excitation were effective.
In Fig. 2(d-f), the effect of adding inhomogeneous conditions is shown. The more significant effect appears in Fig. 2f with the introduction of the coil sensitivity profile, where the right breast emits a higher signal than the left.
In Fig. 3, the displacement of fat pixels with decreasing bandwidth is depicted, a common observation in real measurements.
Finally, in Fig. 4, the phantom used for simulation is interpolated to have overlapping voxels of fat and FGT (Fig. 4a). When simulating for different TEs (Fig. 4b-c), in voxels with both tissues, we find signal variation due to the tissues’ different resonance frequencies. In Fig. 4b, the higher signal intensity reveals fat and water are in phase, while in Fig. 4c the reduced intensity reveals they are opposed. With this work, the potential of MR-zero for breast MRI planning and testing was showcased. Different features were tested, revealing the simulation’s capability of recreating artefacts commonly seen in breast MRI such as fat pixel displacement, fat-water dephasing, and selective saturation/excitation effects. The new extension of the simulation presented in [4], which allows the use of off-resonant RF pulses, was used for the simulation of fat saturation and water excitation FLASH sequences, which produced results according to the expected. In conclusion, simulating realistic breast MRI acquisitions is now an easy extension to the work previously developed with MR-zero which, so far focused on brain MRI. Fast prototyping and testing of new (Pulseq) sequence approaches for MAMMA MRI is now possible with realistic and fast simulation feedback.
Magda DUARTE (Erlangen, Germany), Felix DIETZ, Tobias DORNSTETTER, Jonathan ENDRES, Simon WEINMÜLLER, Sebastian BICKELHAUPT, Moritz ZAIß
15:10 - 15:20
#45718 - PG056 Comparison of Retrospective Ripple Artifact Reduction Techniques for MR Images of Total Hip Arthroplasties.
PG056 Comparison of Retrospective Ripple Artifact Reduction Techniques for MR Images of Total Hip Arthroplasties.
Slice encoding for metal artifact correction (SEMAC) is commonly employed in MRI scans of total hip arthroplasties to mitigate metal artifacts [1]. However, the combination of spectral profiles during image reconstruction can result in ripple artifacts, which may impact diagnostic accuracy near the implants [2,3]. Wahlen et al. have shown with simulated SEMAC images that ripple artifacts exhibit a distinct distribution in the frequency domain [4]. The objective of this work was to compare filtering approaches in Fourier and wavelet domain for ripple artifact reduction in SEMAC images of patients with total hip arthroplasty.
Ripple Artifact Characteristics:
According to MRI simulations by Wahlen et al. [4], ripple artifacts in SEMAC images have distinct spatial frequencies in the range of 0.5-2.0 cm^-1 (Figure 1a).
MR Imaging:
To verify simulated artifact behaviour with real measurements, we acquired images of a phantom that is comparable to the implants on the in-vivo SEMAC images. The implant phantom contained a total hip arthroplasty (titanium stem and cup, cobalt-chromium femoral head; Medacta, Castel San Pietro, Switzerland) embedded in a five-liter plastic container filled with gadolinium-doped water.
Coronal SEMAC images of the phantom were obtained using a 3T MRI scanner (MAGNETOM Prisma; Siemens Healthineers AG, Forchheim, Germany) at Balgrist Campus, Zurich, Switzerland with a 32-channel spine-coil and an 18-channel body-coil. The imaging parameters were TR/TE: 2000/20ms, voxel size: 1.2x1.2mm2, bandwidth: 500Hz/Px, slices: 28, slice thickness: 5mm, SEMAC steps: 16.
Additionally, we processed coronal compressed sensing-based STIR SEMAC images of one patient with total hip arthroplasty. The images were obtained using a 1.5T MRI scanner (MAGNETOM Sola; Siemens Healthineers AG, Forchheim, Germany) at Balgrist University Hospital, Zurich, Switzerland with a 32-channel spine-coil and an 18-channel surface-coil. The imaging parameters were TR/TE: 5000/37ms, TI: 145ms, voxel size: 1.0x1.0mm2, bandwidth: 539Hz/Px, slices: 28, slice thickness: 3.5mm, SEMAC steps: 12) [5]. The corresponding images and Fourier domains (FD) are shown in Figure 1b and c.
Data Post-Processing:
The in-vivo data was filtered in both domains, i.e. Fourier and wavelet domain, and image quality compared for both filtering techniques.
Fourier Domain Filter (FD-Filter):
To filter spatial frequencies related to ripple artifacts, we implemented a FD-Filter (Figure 2a): After applying the Fourier transformation (FT) to the manually drawn Region-of-Interest (ROI) with the artifact, we determined the radius r of the radial spectral energy density (RSED) distribution peak while neglecting low frequency components [4]. A Gaussian ring mask with radius r and adjustable width was then applied to the k-space data within the ROI, followed by an inverse FT.
Wavelet Domain Filter (WD-Filter):
Ripple artifacts exhibit a short wave-like pattern, making the wavelet domain (WD) appropriate for artifact filtering [6](Figure 2b). Three wavelets, Beylkin (beyl), Daubechies (db), and Fejer-Korovkin (fk), were used for the wavelet transformation (WT). Within the ROI, the detail coefficients linked to the ripple artifacts were nulled, using a variable threshold based on the peak WD signal. The filtered image was finally obtained by an inverse WT. Applying the FD-Filter with varying Gaussian ring mask widths shows reduced ripple artifacts (Figure 3). While a larger width reduces more ripple artifacts, it also increases image blurriness. Furthermore, the ROI borders in the image are distinctly noticeable, particularly with the broader masks, which can disrupt the visual coherence of the image. The ROI's FD indicates that the distinct frequency distribution of the ripple artifacts can be effectively removed, however at cost of image quality.
The WD-Filter with different wavelets significantly reduced ripple artifacts with no visible wave-like patterns remaining (Figure 4). The beyl and db wavelets introduced more blurring than the fk wavelet. Artifact removal is also visible in the FT ROI. Both filters can reduce ripple artifacts, but the FD-Filter leaves residual artifacts and compromises image quality, making it unsuitable for clinical use. Precise filtering with optimal radius through RSED distribution is challenging, as the peak is often unclear in in-vivo images due to high k-space intensities overlapping with ripple artifact frequencies. In contrast, the WD-Filter effectively eliminates these artifacts without affecting image quality. We also noted the removal of frequencies corresponding to the artifact in the ROIs FD. Furthermore, individually adjusting the filtering threshold for specific artifacts allows for more adapted corrections. Both approaches can effectively reduce ripple artifacts. However, the WD-filter outperforms the FD-filter in terms of image quality. This filtering method can be particularly beneficial for imaging patients with hip arthroplasties.
Jeanette Carmen DECK (Zurich, Switzerland), Reto SUTTER, Constantin VON DEUSTER
15:20 - 15:30
#45821 - PG057 On the influence of slice profile and flip angle on the signal of frequency-modulated bssfp sequences.
PG057 On the influence of slice profile and flip angle on the signal of frequency-modulated bssfp sequences.
BSSFP sequences offer the highest SNR per unit time of any known MRI sequence, coupled with short scan times. Their signal amplitudes are distinctly influenced by off-resonance and sensitive to alterations in T1, T2, flip angle and TR [1,2]. Various phase-cycled bSSFP approaches [3-5] for quantitative MRI are already exploiting this characteristic off-resonance profile [6]. The signal behavior in the complex plane is described by Eq. (1) – (4) [3-6] in Fig. 1. Thereby, M0 is the equilibrium magnetization, α the flip angle, and θ the spin phase evolution per TR.
Current bSSFP-based mapping approaches require multiple phase-cycled image acquisitions to employ the elliptical signal model. A frequency-modulated (fm) method captures the entire phase cycle in a single measurement [2,7,8] by slowly shifting the phase of the pulse with each acquired k-space line. For slow shifts, the elliptical signal model can closely approximate the fm-bSSFP signal [7,9].
In 2D measurements, the slice profile of the RF pulse can distort the signal, which previously called for improved slice profiles [10]. In contrast, we propose a new fitting model (Fig. 1, Eq. (5)) that accounts for the slice profile by summing up the elliptical signal for N flip angles.
Measurements were performed on a 3T scanner (Magnetom Prisma Fit,Siemens Healthineers, Forchheim, Germany) with a 16-channel head coil, using turkey meat as test specimen with relaxation times similar to human tissue. Reference relaxation times were determined by inversion recovery (T1) and multi-echo spin-echo measurements (T2).
Three Sinc pulses with different durations (RF_dur = 0.9, 2.16, 5.4 ms) and time-bandwidth products (tbp = 3.0, 7.2, 18.0) were applied in fm-bSSFP slice profile measurements and imaging with different flip angles (20°, 40°, 60°).
First, fm-bSSFP signals were simulated using Bloch equations to illustrate the effect of slice profile quality on the complex signal. Then, signals with a range of T1 (300 ms < T1 < 2000 ms) and T2 (10 ms < T2 < 300 ms) were generated from the proposed model, including the measured slice profiles. After adding artificial white noise, the signals were fitted according to Eq. (5) and (1) and the fitted relaxation times were compared to the input. Other settings in this in silico study were similar to the measurements.
2D fm-bSSFP measurements were acquired using tiny golden angle radial sampling with settings: RF_dur = 0.9 ms, tbp = 3.0, RF-Bandwidth = 3.33 kHz, modulation rate = 0.01°/TR, FA = 20°, 40°, 60°, TR = 2*TE = 8.0 ms, resolution = 1.2 x 1.2 mm^2, slice thickness = 10 mm, bandwidth = 980 Hz/px, 36864 spokes, T_AQ = 4:55 min.
Reconstruction included gridding and a sliding window reconstruction with 360 k-space lines per image, resulting in 100 images per phase cycle. The center of the measured signals was rotated to the positive x-axis using parts of the CELF [4] algorithm. Relaxation times and RMSEs from fitting according to Eq. (1) and (5) were compared. Pulse shapes and measured slice profiles are shown in Fig. 2.
Bloch simulations show that considering the slice profile results in severe deviations from the elliptical signal model (Fig. 3(a) and (b)). Fig. 3(a) illustrates that distortion of the signal shape is present for all pulses and strongest for the shortest pulse.
Fig. 4 demonstrates how these distortions corrupt fitting T1 and T2 with the elliptical signal model over the simulated range of T1 and T2. Employing Eq. (1) results in severe systematic over- or underestimation (a-d,i-l). For (Eq. 5), the deviations stay within 10 % of the ground truth. However, deviations in T1 increase with increasing T1 (e-h), and in T2 with increasing T1 and T2 (m-p).
Fig. 3 (c) – (e) show exemplary measured signals from the turkey meat specimen (reference: T1= (803 ± 24.3) ms and T2 = (37.9 ± 0.51) ms). Relaxation times obtained from the elliptical model (Eq. 1) show drastic underestimation for T1 (75 % to 92 %) and a smaller underestimation for T2 (17 % to 19 %). Taking the slice profile into account, T1 is underestimated by 3 % to 25 % and T2 by up to 9 % for the 20° pulse. The RMSEs are significantly smaller (factor 2) for the proposed model (Eq.5). The signal of 2D fm-bSSFP shows clear deviations from the elliptical model (Eq.(1)) due to different flip angles across the slice profile. Model-based relaxometry thus either requires optimized pulses or an improved model, as proposed here. The latter allows a free choice of pulse shape and flip angle, enabling shorter RF pulses, TR and scan time reduction as well as SAR optimization.
Future approaches should eliminate the necessity to measure the slice profiles, for example by working with estimated slice profiles based on the implemented pulse shapes. Furthermore, in vivo validation is necessary. The proposed signal model, which considers the slice profile, provides a more accurate quantification of relaxation times with fm-bSSFP than models ignoring the slice profile.
Clemens MEY (Würzburg, Germany), Hannah SCHOLTEN, Herbert KÖSTLER, Anne SLAWIG
15:30 - 15:40
#47724 - PG058 Simulating how tissue microstructure affects multimodal MRI.
PG058 Simulating how tissue microstructure affects multimodal MRI.
MRI has many different contrast mechanisms that are sensitive to tissue microstructure, including diffusion-weighted MRI, susceptibility-weighted MRI, magnetisation transfer, and quantitative relaxometry. Many of these MRI modalities are sensitive to different aspects of the same microstructural components (e.g., myelin). Thus, combining information across modalities may provide a more comprehensive view of tissue microstructure. However, different modalities are usually analysed in isolation with each one coming with its own set of models and assumptions. Here we present a new Monte Carlo MR (MCMR) simulator[1] that aims to capture different ways in which tissue microstructure can affect the MRI signal evolution for a wide range of MRI sequences (Figure 1).
MCMR simulator was implemented in the Julia programming language[2]. It has both a Julia and command line interface, with comprehensive documentation and tutorials available for both.
An overview of the simulator methodology is shown in Figure 2. Briefly, the user synthesises a tissue geometry (consisting of any combination of infinite walls, infinite cylinders, spheres, and arbitrary meshes), and defines one or more MR sequences for which the MR signal will be computed in parallel. These sequences can include finite or instantaneous radiofrequency (RF) pulses and gradients and can be flexibly defined by the user or directly read from pulseq files[3]. For each sequence the simulator predicts the MRI signal for a single voxel.
The simulator uses a Monte Carlo approach, where for each simulated isochromat we consider (“Tissue Properties” in Figure 1):
1. The isochromat random walk hindered by tissue membranes (which might be permeable).
2. Longitudinal (T1) and transverse (T2) relaxation times, which can vary in individual cells/compartments.
3. The tissue magnetic susceptibility affecting the local magnetic field strength.
4. Surface relaxation and/or magnetisation transfer at the tissue boundary.
All of these features are supported for simplified geometries of cylinders or spheres as well as full meshes. Parameters controlling these effects can be set per cell type, per individual cell, or even for a patch of membrane within a cell.
More details on the methodology can be found in our preprint[1]. Figure 3 illustrates the simulator result for a diffusion MRI sequence and a magnetisation transfer sequence in a substrate made of randomly distributed parallel cylinders. While analytical approximations such as the Gaussian phase approximation[4] for diffusion MRI and the binary spin-bath model[5] for MT give accurate results in certain regimes, they cannot capture all effects, such as the attenuation at high b-values (Figure 3A) or at long mixing times within the free water compartment (red line in Figure 3B). By combining the effects of diffusion, permeability (exchange), magnetic susceptibility, and magnetisation transfer in the MCMR simulator, we allow for a more coherent analysis of how the tissue microstructure affects the MRI signal across different modalities. For example, the simulator could be used to:
• Identify how the various simulated features affect the estimates of modalities where they are not usually considered, such as the effect of permeability and magnetisation transfer on diffusion MRI measurements[6].
• Optimise acquisition protocols for specific aspects of the tissue microstructure[7].
• Model fitting for MR modalities for which no accurate analytical approximations exist, such as MR fingerprinting[8,9].
• Investigate how tissue microstructural changes affect the MRI signal across multiple modalities (Figure 4). This could be used to help interpret changes seen in multi-modal MRI acquisitions. By supporting arbitrary sequences and multiple signal formation mechanisms, the new MCMR simulator[1] enables MR signal prediction across multiple MR modalities and the development of new MR sequences.
Michiel COTTAAR (Oxford, United Kingdom), Zhiyu ZHENG, Karla MILLER, Benjamin C. TENDLER, Saad JBABDI
15:40 - 15:50
#47975 - PG059 k-Space Representation of Arbitrary 3d Shapes for Signal Simulation.
PG059 k-Space Representation of Arbitrary 3d Shapes for Signal Simulation.
MRI signal simulation methods generate synthetic k-space data which can be used for testing new reconstruction methods or for educational purposes. In the past, k-space simulations of $2$d- objects have been integrated into the open-source software BART [1] by using the analytical Fourier transform of geometric objects [2,3,4]. In this work, we extend the simulation framework in BART by providing a generic implementation of the analytical Fourier transform of arbitrary shapes which are described by a triangulation of their surface. This triangulation can readily be obtained from 3D modelling or segmentation software by storing the geometry in the STL fileformat which is the standard fileformat in 3d printing. We demonstrate our simulation by generating k-space for the complex $3$d geometry of a human brain.
The extended BART k-space signal simulation framework generates for each tissue type an individual signal using the usual signal equation in parallel MRI (1) with $z$-component of the field as described by Eq (2). The magnetization function $M$ of one tissue type has a constant value over the geometric area where the tissue is present and tissue-specific properties like relaxation $R2 = 1/T2$ or tissue-dependent off-resonances can be pulled out of the integral (3). One obtains Eq. (4) using the convolution theorem and since the sensitivities are smooth functions, their representation in k-space decays rapidly which leads to Eq. (5).
The surface of the volume $\Omega_M$ is decomposed into $M$ triangles. The segmentation was created with ITK-Snap [5] and containts grey matter, white matter, and CSF of a human brain of a healthy volunteer scanned on a 3T scanner (Siemens Healthineers, Erlangen) from a healthy volunteer with written informed consent. We acquired 3d data with MP2RAGE, MPRAGE and FLASH sequences.
The Fourier transform of the indicator function of the triangulated volume can be simplified to a weighted sum over the Fourier transforms of the triangles over its surface according to Eq. (6). The Fourier transform for
triangles was used in previous work [2,4]. For 3d simulation of the segmented tissue, the magnetization function M in (1) is the indicator function of the triangulated volume. We found that the 3d MP2RAGE sequence with a voxelsize of isotropic 1mm is best suited for segmentation of grey matter, white matter, and CSF (TE 2.96ms, TR 5s, TI$_1$ 996ms, FA$_1$ 4 degree, TI$_2$ 2990ms, $FA_2$ 5 degree, resolution of 256x216x128). Figure 3 shows the resulting segmentations.
For k-space signal simulation, Eq. (1) was evaluated on a 3d Cartesian lattice with matrix size 128x128x128. The resulting single-channel dataset was reconstructed using the 3d inverse discrete Fourier transform and showed typical ringing artifacts as shown in Figures 4 and 5.
The implementation of Eq. (6) was parallelized and the computations were performed on a hardware with 256 cores AMD EPYC 7662 and took ca 4h for an STL file with ca 500000 triangles for 128x128x128 pixel. The simulation of k-space signals for the segmentation volumes that are described by triangles was successfully implemented and the reconstructed images show realistic MRI-specific artifacts such as ringing due to the limited number of high-frequency k-space datapoints.
The generic implementation of Eq. (6) as part of the BART simulation framework enables one to extend the simulation with arbitary k-space sampling trajectories, T2 relaxation, off-resonances, and coil sensitivity encoding as shown in Eq. (5) and described previously [2] and can be included to obtain even more realistic tissue simulation.
The implementation of Eq. (6) can be further improved by making use of GPUs. We extended the BART simulation framework by including the implementation of the analytical Fourier transform of generic shapes whose surface is described by triangulation.
The k-space simulation of STL files obtained by segmentation of MRI images allows a direct comparison of the simulated k-space signals with MRI measurements.
Martin HEIDE (Göttingen, Germany), Martin UECKER
15:50 - 16:00
#47826 - PG060 Variable flip angle chemical shift encoded MRI for PDFF, T1 and R2* estimation in whole body imaging.
PG060 Variable flip angle chemical shift encoded MRI for PDFF, T1 and R2* estimation in whole body imaging.
Whole-body MRI (WB-MRI) is recommended for disease characterisation and treatment response assessment in metastatic cancers such as prostate, breast, and multiple myeloma [1]. A key component is fat-water imaging, with fat fraction maps being shown to outperform apparent diffusion coefficient in detecting early response in myeloma [2].
International guidelines for both advanced prostate cancer and myeloma recommend acquisition of a 2-point T1-weighted Dixon sequence as a compromise for combined morphological imaging and fat fraction estimation [3]. However accurate estimation of proton density fat fraction (PDFF) requires correction for several sources of error [4-9].
Fat quantification in bone marrow is sensitive to T1 bias, owing to the large difference in the T1 of the water and fat compartments [10]. Variable flip angle chemical shift encoded (VFA-CSE) MRI has been shown to minimize T1 bias in comparison to low flip angle approaches [11]. There is also potential for T1 to be a potential clinical biomarker for disease response.
This study aimed to develop whole-body VFA-CSE MRI for accurate quantification of PDFF and T1 in bone marrow malignancies in the presence of significant T1 variation.
An in-house pulse sequence based on a 3D RF-spoiled gradient echo was used for VFA-CSE MRI. The manufacturer’s 6-point Dixon sequence from the LiverLab package was also run for comparison [12].
T1 validation was performed on the same scanner with using a relaxometry phantom [13]. B1+ maps were acquired with a Bloch-Siegert pulse sequence [14] to correct for transmit field inhomogeneities for the VFA-CSE approach. The nominal T1 values provided by the manufacturer of the vials were used as the reference without temperature correction.
Volunteer whole-body scanning was performed as a proof of concept. The volunteer provided written consent to participate in the study.
Validation of PDFF with MRS STEAM at 3T (for superior spectral separation) used a peanut oil phantom based on Zhao et al [15]. MRS was treated as the reference standard. Processing was performed in jMRUI using AMARES [16], with T2 correction prior to calculation of PDFF using a 7-peak model of bone marrow [17].
Details of sequence parameters are in Figure 1.
Reconstructions were done in Python (3.12). A schematic flowchart of the approach is shown in Figure 2 [18].
To assess the value of including VFA in the signal model for PDFF estimation, the fit was completed with and without the second pass/joint fitting step.
LiverLab data were reconstructed with the standard inline reconstruction, which assumes a fat spectrum of the human liver at body temperature.
Comparisons for both T1 and PDFF with their respective gold standards were performed using Bland-Altman analysis.
Volunteer data were acquired in five stations from skull vertex to mid-thigh and reconstructed to generate whole-body quantitative maps. The proposed VFA-CSE MRI PDFF showed better agreement with MRS estimates than LiverLab in the phantom, as shown below in Figure 3. The vial with pure peanut oil was underestimated by VFA-CSE MRI compared to MRS (90.25% vs 100%). Considering only the first pass of VFA-CSE (PD-CSE), there is a nonlinear bias in PDFF, highlighting the importance of T1 correction. Similarly, inclusion of B1+ estimation into VFA-CSE reduces bias in T1 estimation from 14.0% to 6.5%.
Coronal and sagittal reformats from the volunteer reconstruction are shown in Figure 4. In this work, we have demonstrated VFA-CSE as a promising approach for PDFF and T1 estimation in a whole-body context. Superior agreement with spectroscopy can be achieved in tissues with large T1 disparities between fat and water, such as bone marrow, compared to a conventional PDFF reconstruction. The performance of the inline LiverLab reconstruction likely reflects the incorrect assumption of body temperature, which is known to be a significant confounder for magnitude-based fitting [19].
VFA-CSE MRI underestimated PDFF in the pure fat vial, though this fat fraction is not truly clinically relevant for treatment response in malignant bone disease [20].
The potential impact of partial spoiling was not modelled, which may explain part of the discrepancy in T1 following B1+ correction in the NIST phantom alongside temperature.
For T1 to be used as an imaging biomarker, B1+ correction is necessary, particularly given the left-right variation in T1 seen in the volunteer’s pelvis and shoulders in Figure 2. Estimating it through signal modelling may require a third pass, as demonstrated by Roberts et al in liver [21]. This would necessitate an increase in the number of breath-holds or a reduction in the achievable resolution. Future work will investigate data-driven methods of B1+ estimation. We have demonstrated a VFA-CSE acquisition and reconstruction for whole-body imaging in malignant bone disease, which corrects for T1 bias in PDFF measurements as well as providing T1 and R2* maps.
Azma YASSINE (London, United Kingdom), Pete J. LALLY, David J. COLLINS, Nina TUNARIU, Dow-Mu KOH, Christina MESSIOU, Geoff CHARLES-EDWARDS, Christina TRIANTAFYLLOU, Neal K. BANGERTER, Jessica M. WINFIELD
|
Salle Major |
16:00 |
TIME FOR A BREAK
- Coffee and refreshments will be available at the cash bar.
|
16:30 |
"Saturday 11 October"
Added to your list of favorites
Deleted from your list of favorites
A36
16:30 - 18:00
MANSFIELD LECTURE & CLOSING
16:30 - 18:00
It’s the Magnet, stupid!
Oliver SPECK (Keynote Speaker, Magdeburg, Germany)
|
Auditorium 900 |
18:00 |
"Saturday 11 October"
Added to your list of favorites
Deleted from your list of favorites
A37
18:00 - 19:00
Business meeting
|
Auditorium 900 |
19:00 |
CONGRESS DINNER
|