Novel & Multicontrast Approaches
Contrast Mechanisms Monday, 17 May 2021
Digital Poster
1232 - 1251

Oral Session - Novel & Multicontrast Approaches
Contrast Mechanisms
Monday, 17 May 2021 14:00 - 16:00
  • Comparison of inhomogeneous Magnetization Transfer (ihMT), R1 and MPF for myelin specific imaging
    Andreea Hertanu1,2, Lucas Soustelle1,2, Arnaud Le Troter1,2, Julie Buron1,2,3, Julie Le Priellec3, Myriam Cayre3, Pascale Durbec3, Gopal Varma4, David C. Alsop4, Olivier M. Girard1,2, and Guillaume Duhamel1,2
    1Aix Marseille Univ, CNRS, CRMBM, Marseille, France, 2APHM, Hôpital Universitaire Timone, CEMEREM, Marseille, France, 3Aix Marseille Univ, CNRS, IBDM, Marseille, France, 4Division of MR Research, Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
    IhMT filtered from short-T1D components and corrected for T1/B1 effects (ihMTsat) offers the highest specificity to healthy myelinated tissues in the mouse brain when compared to other metrics such as MPF, T1 and ihMTR.
    Figure 2: Representative slices of the 3D templates for R1, MPF, ihMTR, ihMTsat with different T1D-filtering along with plp-GFP fluorescence microscopy images at -3.2, -0.7 and +0.7 mm from bregma. Brain structures in WM (IC – internal capsule, CC – medial corpus callosum, ON – optical tract), Grey Matter (CTX – cerebral cortex, HP – hippocampus) and mixed WM/GM structures (TH – thalamus, CP – caudoputamen) where quantitative analyses were performed are indicated on the T2w/plp-GFP images.
    Figure 3: Correlations of MR metrics and plp-GFP signal. a) Pearson correlation coefficient matrix for all techniques. b) Linear regressions of normalized R1, MPF, ihMTR0.8 and ihMTsat0.8 with GFP. c) Linear regression of MPF with ihMTsatCM, ihMTsat0.8, ihMTsat1.6 and ihMTsat3.2. Shaded areas correspond to confidence curves for line fits with an α-level of 0.1.
  • Uncovering the specificity of quantitative MRI to different molecular forms of iron in the brain.
    Shir Filo1, Rona Shaharabani1, and Aviv Mezer1
    1The Edmond and Lily Safra Center for Brain Science, The Hebrew University of Jerusalem, Jerusalem, Israel
    We propose an in vivo quantitative MRI approach for assessment of iron forms, based on the dependency of R1 on R2*. Our method was established in phantoms and validated against histology. It predicts the heterogeneous distribution of iron-binding proteins with age and across the in-vivo brain.
    Establishing the iron relaxivity framework in vitro. (a) R2* and R1 vary with the concentration of iron-binding proteins (x-axis), with their type (transferrin, ferritin) and with the molecular environment (liposomes, saline, BSA) (different colors). Data points are phantom samples’ medians. Symbols represent water fractions. (b) The dependency of R1 on R2* for different molecular compounds and environments of iron. R2* and R1 of samples with varying water and protein concentrations were binned, data points are the bins’ median. The R1-R2* slopes are different for each iron form.
    Fully-constrained model predicts the fractions of iron-binding proteins in the in vivo human brain. The measured R1-R2* slope in each brain area was modeled (Eq. 1) as a weighted sum of the R1-R2* slopes of transferrin (Tf) and ferritin (Fer) (estimated in liposomal phantoms). The Tf and Fer fractions sum to one (Eq. 2). Rearranging the model (Eq. 3) allows to predict the transferrin fraction (Tf/(Tf+Fer), y-axis) for younger (<64) and older subjects in 11 brain regions. There are no free parameters. In the x-axis is the Tf fraction measured post-mortem5,6,9,10. MAE=mean absolute error.
  • Improved spin-lock based detection of ultra-low-field electro-magnetic oscillations for direct fMRI
    Maximilian Gram1,2, Markus Dippold2, Daniel Gensler1,3, Martin Blaimer4, Peter Nordbeck1,3, and Peter Michael Jakob2
    1Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany, 2Experimental Physics 5, University of Würzburg, Würzburg, Germany, 3Comprehensive Heart Failure Center (CHFC), University Hospital Würzburg, Würzburg, Germany, 4Fraunhofer Institute for Integrated Circuits IIS, Würzburg, Germany
    Recently, a spin-lock-based technique for directly measuring neuronal activity was introduced, which demonstrated imaging of alpha activity. In the present work it is shown that the choice of the preparation parameters is of essential importance and enables significant signal increases.
    Figure 3) Simulation results of the parameter variation of tSL and fSL. The resulting NEMO-amplitude a is shown as a heat map (a). In (b) and (c) profiles for tSL and fSL are shown. In (d) the corresponding sinusoidal fits can be seen for varying phases ϕ. The optimal sequence parameters are tSL=82.4ms and fSL=10.625Hz and thus deviate significantly from the parameters chosen in [3] (125ms, 10Hz). In the simulation, typical relaxation times for gray matter T=78ms [3] and T=1.5*T1ρ were considered.
    Figure 5) Results of the parameter variation. In (a) fSL was varied (constant tSL=100ms). Similar to the simulation, the NEMO-amplitude shows a peak at fSL≈10Hz. In (b) tSL was varied (constant fSL=10Hz). The emergence of local maxima can be seen. However, the behavior of RSS(tSL) is remarkable. In the case ≈25ms, ≈75ms and ≈125ms, image artifacts lead to great difficulties in the sinusoidal fit. A reliable determination of the NEMO-amplitude was therefore not possible in this range.
  • Noninvasive Detection of Changes in Membrane Potential with MR Measurements
    Kyeongseon Min1, Sungkwon Chung2, Phan Tan Toi3,4, Jongho Lee1, Seung‐Kyun Lee3,4,5, and Jang-Yeon Park3,4
    1Laboratory for Imaging Science and Technology, Department of Electrical and Computer Engineering, Seoul National Univeristy, Seoul, Korea, Republic of, 2Department of Physiology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, Korea, Republic of, 3Department of Biomedical Engineering, Sungkyunkwan University, Suwon, Korea, Republic of, 4Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, Korea, Republic of, 5Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, Korea, Republic of
    In this study, we observed that depolarization of SH-SY5Y cells leads to an increase in T1 and T2 and a decrease in pool size ratio (PSR). Contrarily, when SH-SY5Y cells were hyperpolarized, T1 and T2 decreased and PSR increased.
    Figure 2. The relative changes in T1 and the corresponding membrane potentials. The dotted black lines in each plot represent the normal condition. (a): Relative T1 changes and the membrane potentials when those are altered by [K+]. (b): Relative T1 changes and the membrane potentials when those are altered by [Ba2+]. (c): Relative T1 changes in (a) and (b) are illustrated on the same plot using the abscissa of membrane potentials.
    Figure 3. The relative changes in T2 and the corresponding membrane potentials. The dotted black lines in each plot represent the normal condition. (a): Relative T2 changes and the membrane potentials when those are altered by [K+]. (b): Relative T2 changes and the membrane potentials when those are altered by [Ba2+]. (c): Relative T2 changes in (a) and (b) are illustrated on the same plot using the abscissa of membrane potentials.
  • High temporospatial resolution MR imaging of neuronal activity in vivo
    Phan Tan Toi1,2,3, Hyun Jae Jang4, Jeehyun Kwag4, and Jang-Yeon Park1,2,3
    1Department of Biomedical Engineering, Sungkyunkwan University, Suwon, Korea, Republic of, 2Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, Korea, Republic of, 3Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, Korea, Republic of, 4Department of Brain and Cognitive Engineering, Korea University, Seoul, Korea, Republic of
    In vivo direct MR imaging of neuronal activity at a high temporospatial resolution
    Figure 2. Temporospatial imaging of neuronal activity propagation. (a) Illustration of DIANA-fMRI experiment with a coronal slice containing both thalamus and S1BF activated by electrical stimulation applied to mouse whisker pad. (b) DIANA time series from thalamus, contralateral S1BF (n = 10). Dotted line indicates the stimulation onset. (c) Time series of t-value maps for 30 ms after stimulation with 5 ms temporal resolution in one representative mouse (paired t-test, p < 0.05, cluster size > 5 voxels). Dashed circular markers highlight the DIANA response areas. Error bar: S.E.M.
    Figure 3. In vivo electrophysiological validation of neuronal activity. (a) Illustration of simultaneous electrophysiological recording in vivo both thalamus and contralateral S1BF. (b) Representative MUA signals and single-unit spikes acquired in thalamus (top) and contralateral S1BF (bottom). (c) Post-stimulation time histogram of the responsive single units over time plotted with DIANA signals in thalamus (top, 7 units from n = 7) and contralateral S1BF (bottom, 16 units from n = 7). Dotted lines indicate the stimulation onset. Shaded area: S.E.M.
  • Diffuse axonal injury has a specific multidimensional MRI signature in traumatically injured corpus callosum
    Dan Benjamini1, Diego Iacono2, Michal Komlosh1, Daniel Perl2, David Brody2, and Peter Basser1
    1National Institute of Child Health and Human Development, Bethesda, MD, United States, 2Uniformed Services University of the Health Sciences, Bethesda, MD, United States
    Can microscopic traumatic axonal injury be imaged noninvasively? We show that multidimensional MRI, which combines T1 , T2 and diffusion, can reveal damage that is invisible using quantitative MRI modalities, and provide an injury-only image that visualizes microscopic lesions in the brain.
    Summary of the findings, illustrated by comparing the FA with the T1-T2 injury image.
    APP density (% area) from 132 tissue regions, consisting of 4 APP-positive regions from each TAI case (total of 32, blue dots), 4 to 6 normal-appearing WM regions from all cases (total of 56, red dots), and 4 cortical GM regions from all cases (total of 44, yellow dots), and the corresponding MR parameter correlations. Individual data points represent the mean ROI value from each post-mortem tissue sample. Scatterplots of the mean (with 95% confidence interval error bars) % area APP and all MR parameters.
  • Single-shot simultaneous diffusion and T2 mapping based on overlapping-echo detachment planar imaging
    Lingceng Ma1, Xinran Chen1, Jian Wu1, Lijun Bao1, Shuhui Cai1, Congbo Cai1, and Zhong Chen1
    1Department of Electronic Science, Xiamen University, Xiamen, China
    A single-shot method through overlapping-echo detachment planar imaging (DT2M-OLED) was proposed for simultaneously delivering diffusion and T2 mapping within around one hundred milliseconds.
    Figure 3. In-vivo rat brain experimental results. The rats were under deep anesthetic. Four regions of interest (ROI) were chosen on each rat brain for analysis, which are marked on SE-EPI images.
    Figure 4. In-vivo rat brain experimental results. The rat was recovering from deep anesthetic during acquisition. (a) Reference ADC maps from SE-EPI. (b) Reference T2 maps from SE-EPI. (c) Temporal ADC maps from DT2M-OLED. (d) Temporal T2 maps from DT2M-OLED.
  • Deep learning based acceleration of multi-contrast MRI examinations by acquiring contrast and sharing inter-contrast structure information
    Sudhanya Chatterjee1, Suresh Emmanuel Joel1, Sajith Rajamani1, Shaik Ahmed1, Uday Patil1, Ramesh Venkatesan1, and Dattesh Dayanand Shanbhag1
    1GE Healthcare, Bangalore, India
    We used deep learning to accelerate MRI examinations where multiple contrasts (T2FSE, FLAIRT1, FLAIRT2) are acquired for the same subject using a contrast preserving and structure sharing approach.
    Figure-3 Accelerated FLAIR T1 and FLAIR T2 images (at 2x and 3x) are shown for a test case. For this MR examination, all three contrasts were acquired on a 1.5T GE Signa Creator MR system. The FLAIR (accelerated scans) and T2FSE (reference scan) were obtained at same in-plane resolution but different slice thickness and slice spacing (FLAIR T2 being thicker). The proposed method is designed to deal with such acquisitions. All the accelerated reconstructions have high SSIM values showing robust and high quality reconstruction
    Figure-2 The training workflow is shown here. The network is trained on both reference image and image obtained from the grafted k-space. It predicts the grafting artifact which is then removed from the accelerated image to obtain a clean image
  • MULTI-Parametric MR imaging with fLEXible modular design (MULTIPLEX)
    Yongquan Ye1, Jingyuan Lv1, Yichen Hu1, Zhongqi Zhang2, Jian Xu1, and Weiguo Zhang1
    1UIH America, Inc., Houston, TX, United States, 2United Imaging Healthcare, Shanghai, China
    Multi-parametric MR imaging method with flexible modular design
    Figure.3 Examples of MULTIPLEX’s calculated images of the same data.
    Figure.1 Exemplary design of the MULTIPLEX sequence, which directly generates from k-space 2(N1+N2) sets of images.
  • R2* mapping of the whole brain with 0.8 mm isotropic resolution at 7T in less than 7 minutes
    Arun Joseph1,2,3, Tobias Kober4,5,6, and Tom Hilbert4,5,6
    1Advanced Clinical Imaging Technology, Siemens Healthcare AG, Bern, Switzerland, 2Translational Imaging Center, Sitem-Insel, Bern, Switzerland, 3Departments of Radiology and Biomedical Research, University of Bern, Bern, Switzerland, 4Advanced Clinical Imaging Technology, Siemens Healthcare AG, Lausanne, Switzerland, 5Department of Radiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland, 6LTS5, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
    We implemented a compressed sensing multi-echo GRE acquisition at 7T to acquire 0.8 mm isotropic R2* maps of the whole brain in <7 mins. A reduction in susceptibility artifacts is observed in comparison to acquisitions with 1.6 mm isotropic resolution.
    Figure 2: R2* maps from different views obtained from GRAPPAx2 and compressed sensing reconstructions with 1.6 mm and 0.8 mm isotropic resolutions.
    Figure 5: Comparison of sagittal R2* maps obtained from GRAPPAx2 and CS acquisition with 1.6 mm and 0.8 mm resolution.
Back to Top
Digital Poster Session - Multicontrast Methods
Contrast Mechanisms
Monday, 17 May 2021 15:00 - 16:00
  • Quantitative myelin-sensitive MRIs exhibit differential sensitivity to multiple sclerosis pathology in distinct brain lesions and regions
    Reza Rahmanzadeh1,2,3, Po-Jui Lu1,2,3, Muhamed Barakovic1,2,3, Matthias Weigel1,2,3, Laura Gaetano4, Riccardo Galbusera1,2,3, Thanh D. Nguyen5, Francesco La Rosa 6,7, Daniel S. Reich8, Pascal Sati8,9, Yi Wang5, Meritxell Bach Cuadra6,7, Ernst-Wilhelm Radue1,2, Jens Kuhle1,3, Ludwig Kappos1,3, Stefano Magon10, and Cristina Granziera1,2,3
    1Neurologic Clinic and Policlinic, Departments of Medicine, Clinical Research and Biomedical Engineering, University Hospital Basel and University of Basel, Basel, Switzerland, 2Translational Imaging in Neurology (ThINk) Basel, Department of Biomedical Engineering, University Hospital Basel and University of Basel, Basel, Switzerland, 3Research Center for Clinical Neuroimmunology and Neuroscience (RC2NB) Basel, University Hospital Basel and University of Basel, Basel, Switzerland, 4Hoffmann-La Roche Ltd., Basel, Switzerland, 5Department of Radiology, Weill Cornell Medical College, New York, NY, United States, 6Signal Processing Laboratory (LTS5), Ecole Polytechnique Fédérale de Lausanne (EPFL), Laussane, Switzerland, 7Radiology Department, Center for Biomedical Imaging (CIBM), Lausanne University and University Hospital, Laussane, Switzerland, 8Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, NIH, 10 Center Drive MSC 1400, Building 10 Room 5C103, Bethesda, MD, United States, 9Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, United States, 10Pharmaceutical Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
    Quantitative T1, myelin water imaging and quantitative susceptibility mapping (QSM) show differential sensitivity to multiple sclerosis pathology in white matter and cortical lesions, peri-plaque and normal appearing tissue.
    Figure 2. Voxel-wise randomized clustering comparison between NAWM patients and WM controls in A) qT1, B,C) QSM and D) MWF and susceptibility, respectively.
    Figure 3. Vertex-wise comparison between NAGM patients and GM controls in A) qT1, B,C) QSM and D) MWF, respectively.
  • Three-dimensional whole-brain simultaneous quantitative mapping of T1, T2, T2*, and susceptibility with MR Multitasking
    Tianle Cao1,2, Sen Ma1, Nan Wang1, Sara Gharabaghi3, Yibin Xie1, Zhaoyang Fan1,4,5, Elliot Hogg6, E. Mark Haacke 3,7,8, Michele Tagliati6, Anthony G. Christodoulou1,2, and Debiao Li1,2
    1Biomedical Imaging Research Institute, Cedars Sinai Medical Center, Los Angeles, CA, United States, 2Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, United States, 3Magnetic Resonance Innovations, Inc., Bingham Farms, MI, United States, 4Department of Radiation Oncology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States, 5Department of Radiology, University of Southern California, Los Angeles, CA, United States, 6Department of Neurology, Cedars Sinai Medical Center, Los Angeles, CA, United States, 7Department of Radiology, Wayne State University School of Medicine, Detroit, MI, United States, 8The MRI Institute for Biomedical Research, Bingham Farms, MI, United States
    A new approach for simultaneous quantitative mapping of T1, T2, T2*, and susceptibility was developed. Results of both visual comparison and statistical analysis showed that our proposed method agreed well with reference methods while being more time efficient.
    Figure 2. Representative T1/T2/T2*/QSM mapping at 3 slice locations using MR Multitasking and the corresponding reference protocols on a healthy volunteer.
    Figure 4. Results from a 23-year-old female volunteer including qualitative images and quantitative data. The first row shows quantitative maps and the second and third rows show the synthesized weighted images. SWI and tSWI were minimum intensity projection (mIP) results with an effective slab thickness of 16 mm. χ1=0, χ2=450ppb was adopted for tSWI23.
  • 3D Isotropic-resolution Non-rigid Motion Compensated Liver T1, T2 and fat fraction mapping
    Giorgia Milotta1, Gastao Cruz2, Radhouene Neji2, Claudia Prieto2, and Rene Botnar2
    1University College London, London, United Kingdom, 2King's College London, London, United Kingdom
    The proposed approach permits the acquisition of 3D free-breathing T1, T2 and fat fraction maps in a scan time of less than 6 minutes. Increased sharpness and reduced spatial variability was observed with non-rigid motion correction.
    Figure 3 – Co-registered T1 and T2 maps, fat fraction and B0 map for one healthy subject acquired with 3-point Dixon GRE read out. Coronal, sagittal and transversal views are shown. T1=704±30ms, T2 = 58±3ms and fat fraction = 3.3±1.7% were measured within a ROI in the liver. Acquisition parameters included FA=8deg, isotropic resolution of 2mm3, FOV=320x320x168mm3, coronal orientation, 14 echoes for iNAV acquisition, acquisition window of 200ms, bandwidth=801Hz/pixel, T2prep=50ms, TI=120ms and total scan time of ~6min.
    Figure 1 – Four interleaved volumes are acquired with variable density Cartesian trajectory, three-point Dixon GRE readout. 2D-iNAVs are acquired to correct for translational motion and respiratory binning. Water images are generated with Dixon water/fat separation and used to obtain the signal evolution of each voxel. T1 and T2 maps are obtained by matching the acquired signal evolution to the EPG simulated dictionary. Fat fraction and M0 map are obtained from water/fat separation algorithm.
  • Multi-contrast MRI Atlas of the Cynomolgus Macaque Brain
    Rakshit Dadarwal1,2 and Susann Boretius1,2
    1Functional Imaging Laboratory, German Primate Center, Göttingen, Germany, 2Georg August Universität Göttingen, Göttingen, Germany
    We provide a high-resolution multi-contrast MRI template space for the cynomolgus macaque brain in the stereotaxic space. Template space also offers cortical, subcortical, and white matter structural parcellation. 
    Figure 1. The symmetric cynomolgus macaque brain templates in coronal, sagittal, and axial view. A) Templates of the originally acquired weighted data sets (from top to bottom) include T1-weighted, MT-weighted, ME-GRE mean across echo times (mGRE), and T2-weighted template. B) Parametric map templates (from top to bottom) incorporate QSM, R2*, and MT saturation and apparent T1 maps.
    Figure 2. Cynomolgus macaque brain structural parcellation derived from the openly available rhesus macaque brain templates. Emanated ROI labels are from the Cortical Hierarchy Atlas of the Rhesus Macaque (CHARM), D99, Subcortical Atlas of the Rhesus Macaque (SARM), and NeuroMaps atlas.
  • Generation of co-registered multi-contrast MR images for carotid atherosclerosis evaluation based on a single SIMPLE sequence
    Jiaqi Dou1, Yajie Wang1, Huiyu Qiao1, Zhensen Chen1, Yuze Li1, Haikun Qi2, Jie Sun3, Dongxiang Xu3, Xihai Zhao1, and Huijun Chen1
    1Center for Biomedical Imaging Research, School of Medicine, Tsinghua University, Beijing, China, 2School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom, 3Department of Radiology, University of Washington, Seattle, WA, United States

    This study generated a set of co-registered T1w, T2w, PDw images, and MRA based on a SIMPLE sequence. Preliminary experiments validated the feasibility of the generated multi-contrast images for carotid plaque assessment and comparable performance with conventional sequences.

    Figure 1 A, Sequence diagram of SIMPLE, in which three alternate T2-prep pulses of 25, 50, and 0 ms and IR pulses, incorporated with 3D golden-angle radial acquisition, are adopted. B, Simulated signal evolutions of the vessel wall and blood within three continuous shots with different T2-prep pulses. And the illustration of view sharing and sliding window reconstruction of the generated multi-contrast images from SIMPLE, including MRA, T1w, T2w and PDw images. C, Scan parameters of SIMPLE and conventional multi-contrast MR imaging.
    Figure 2 An example of the generated MRA as well as axial MRA, T1w, T2w, and PDw images by SIMPLE from one patient with atherosclerotic plaques, with comparison to the conventional sequences. Coronal maximum intensity projection (MIP) of MRA demonstrated clear depictions of bilateral carotid arteries. IPH (red arrow) showed hyperintensity on both SIMPLE-T1w image and T1w TSE image. Blue arrows indicated the hypointensity of calcification.
  • Multiparametric MRI Distinguishes Cerebral Radiation Necrosis vs. Recurrent Glioma in Mouse Models
    Xia Ge1, John A Engelbach1, Liya Yuan2, Sonika Dahiya3, Feng Gao4, Keith M Rich2, Joseph JH Ackerman1,5,6,7, and Joel R Garbow1,5
    1Radiology, Washington University in St Louis, St Louis, MO, United States, 2Neurosurgery, Washington University in St Louis, St Louis, MO, United States, 3Neuropathology, Washington University in St Louis, St Louis, MO, United States, 4Surgery, Washington University in St Louis, St Louis, MO, United States, 5Alvin J Siteman Cancer Center, Washington University in St Louis, St Louis, MO, United States, 6Internal Medicine, Washington University in St Louis, St Louis, MO, United States, 7Chemistry Department, Washington University in St Louis, St Louis, MO, United States
    A quantitative, clinically translatable 1H MR imaging pipeline has been developed and demonstrated to hold considerable promise for differentiating recurrent tumor from treatment effects (e.g., late time-to-onset necrosis) for high-grade tumor patients treated with radiation.
    Figure 3. The Mixed Model. A) H&E histology; (B) R1PC map; (C) ADC map; (D) T1WPC; (E) MTR map; (F) DCEAUC map from a slice through the brain of a C57BL/6 mouse irradiated hemispherically with a GK into which GL261 tumor cells were orthotopically implanted 4 weeks post irradiation. Images and histology were collected 18 days post tumor implantation. Regions of pathology (tumor or RN) appear hyperintense on R1PC map, ADC map, T1WPC, and DCEAUC map, while only the tumor is hypointense on the MTR map. The orange-colored oval guides the eye to the whole lesion region, yellow to tumor, cyan to RN.
    Figure 4: Box plot of MRI parameter means of lesion (tumor/RN) in the mixed, tumor alone and RN alone models. A. R1; B.R1PC; C. R2; D. ADC; E. MTR; F. DCEAUC. Lesion ROIs were identified by histology. Differences in values of R1, R2, ADC, MTR, DCEAUC, for tumor vs. RN in the mixed model are highly statistically significant. Tumor in tumor alone (n=9) and RN in RN alone (n=9), differences in values of R1, R1PC, R2, ADC, MTR are statistically significant. *p < 0.05, **p < 0.01, ****p<0.0001.
  • Changes of native T1, T1ρ, and T2 values during liver fibrosis in rats at 11.7T MRI
    Yimei Lu1, Qianfeng Wang2, and Dengbin Wang1
    1Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, shanghai, China, 2Fudan University, Institute of Science and Technology for Brain-Inspired Intelligence, shanghai, China
    Our study was to investigate the repeatability of the quantitative parameters at 11.7T, and then to explore the changes of the quantitative parameters in liver fibrosis induced by BDL and injection of CCl4, and to evaluate the performance of quantitative parameters for staging liver fibrosis.
    Fig 3. Images show MRI measurements in vivo for BDL and CCl4 model of liver fibrosis. Representative axial images of (a) native T1 maps, (b) T1 ρ maps and (c) T2 maps for each group (Metavir stages F0-F4). The images (d-f) show the corresponding data for all rats in BDL and CCl4 model. One-way ANOVA followed by Scheffe post hoc test was performed. *P < 0.05, **P < 0.01, ***P < 0.001.
    Fig 1. Images show liver specimens characterization of changes in fibrosis (Metavir stages F0-F4) in BDL and CCl4 model. Representative images of H&E-stained (top row) and Sirius red-stained (bottom row) liver tissue from each experimental group. H&E, hematoxylin and eosin, 200×original magnification; Sirius red stain, 200×original magnification. CCl4, carbon tetrachloride; BDL, bile duct ligation
  • Multi-Physics Multi-Contrast Magnetic Resonance Imaging
    Mehdi Sadighi1, Mert Şişman1, and B. Murat Eyüboğlu1
    1Electrical and Electronics Eng., Middle East Technical University (METU), Ankara, Turkey
    A Diffusion-Weighted (DW) Spin Echo (SE) based pulse sequence with current injection is proposed to acquire multi-contrast MR data based on different physical properties.
    Figure 1: The schematic diagram of the SE based Multi-contrast pulse sequence. gd’s are the magnitudes of the diffusion encoding gradients. I± and TC are the amplitude and the duration of the injected current, respectively. The current pulse is injected after 90° RF pulse until the beginning of gd.
    Figure 5: The reconstructed ECDR (η) and conductivity tensor distributions of the imaging phantom using the vertical and horizontal current injections. (a) η (b) cxx (c) cyy (d) czz. The mean values of the reconstructed anisotropic conductivity for the left and right inhomogeneities are: cxx,Left = 0.33 S/m, cyy,Left = 0.28 S/m, czz,Left = 0.31 S/m and cxx,Right = 0.30 S/m, cyy,Right = 0.32 S/m, czz,Right = 0.35 S/m.
  • Optimising multi-contrast MRI experiment design using concrete autoencoders
    Chantal Tax1,2, Hugo Larochelle3, Joao P. De Almeida Martins4, Jana Hutter5, Derek K. Jones2, Maxime Chamberland2, and Maxime Descoteaux6
    1Image Sciences Institute, University Medical Center Utrecht, Utrecht, Netherlands, 2CUBRIC, Cardiff University, Cardiff, United Kingdom, 3Google Brain, Montreal, QC, Canada, 4Department of Radiology and Nuclear Medicine, St. Olav's University Hospital, Trondheim, Norway, 5Centre for Medical Engineering, King's College London, London, United Kingdom, 6SCIL, University of Sherbrooke, Sherbrooke, QC, Canada
    Multi-contrast MRI provides a comprehensive picture of tissue microstructure, but the high dimensionality of the parameter space increases scan time. In this work, we present a data-driven approach to multi-contrast MRI experiment design using unsupervised machine learning.
    Sampling for both databases, varying inversion time (TI), delay time (TD), (θ,φ), b-value, and b parameterising the ”shape” of the diffusion-encoding b-tensor16 (represented by the ellipsoids, with corresponding encoding waveforms along each axis). Left: images are scaled per volume except in the TD/TE dimension. Right: number of directions for each parameter setting.
    Mean-squared error across voxels, K is the maximum number of selected measurements. Top: distributions for varying K and for each subject left out of the training and feature selection. Bottom: MSE map for subject 1. K= 50 shows a larger MSE in frontal, occipital, and deep gray matter structures.
  • A Semi-Supervised Learning Framework for Jointly Accelerated Multi-Contrast MRI Synthesis without Fully-Sampled Ground-Truths
    Mahmut Yurt1,2, Salman Ul Hassan Dar1,2, Berk Tinaz1,2,3, Muzaffer Ozbey1,2, Yilmaz Korkmaz1,2, and Tolga Çukur1,2,4
    1Department of Electrical and Electronics Engineering, Bilkent University, Ankara, Turkey, 2National Magnetic Resonance Research Center, Bilkent University, Ankara, Turkey, 3Department of Electrical and Computer Engineering, University of Southern California, Los Angeles, CA, United States, 4Neuroscience Program, Aysel Sabuncu Brain Research Center, Bilkent University, Ankara, Turkey
    We propose a semi-supervised model for mutually accelerated multi-contrast MRI that enables synthesis of fully-sampled target images without demanding large datasets of costly fully-sampled source or ground-truth target acquisitions. 
    Fig. 1: a) Supervised methods learn a source-to-target mapping using costly datasets of fully-sampled images of the source- and target-contrasts. b) The proposed semi-supervised model can be trained to synthesize fully-sampled target images using only undersampled ground-truth acquisitions of the target-contrast and allows recovery from undersampled sources to further reduce data requirements. c) The proposed method involves a selective loss function measuring the error only on the acquired k-space coefficients in terms of k-space, image-domain L1 and adversarial losses.
    Fig. 2: Representative results from the ssGAN and fsGAN models are displayed for the synthesis tasks in the IXI dataset with fully-sampled source images (Rsource=1): a) T1-weighted image synthesis and b) T2-weighted image synthesis. Synthesized target-contrast images are displayed for ssGAN-2 (Rtarget=2), ssGAN-3 (Rtarget=3), ssGAN-4 (Rtarget=4), and fsGAN together with fully-sampled reference images.
  • Multi-Contrast Whole Brain MRI: Optimization of Imaging Parameters and Motion Compensation
    Jing Liu1, Angela Jakary1, Duan Xu1, and Janine Lupo1
    1Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, United States
    Optimization of the imaging parameters and motion compensation for multi-contrast whole brain MRI in a single 3D acquisition resulted in imporoved image quality and will benefit for better accuracy of the resulting brain tissue segmentation and lesion detection. 
    Figure 1. a) IIR-bSSFP acquisition. A nonselective inversion pulse is applied followed by a fixed number of segmented bSSFP acquisitions (interval of Tinv). b) With a continuous data acquisition, a series of 3D images are reconstructed at different TIs. c) Simulated signal evolutions of different brain tissues with assumed T1/T2 values and scan parameters (Tinv=3s, flip angle=30 degrees, TR=4ms).
    Figure 3. Whole brain multi-contrast MRI acquired in a single scan of 6 minutes. The imaging sequence has inherent motion robustness to small amounts of motion, but any excess motion would cause image quality degradation without motion compensation (right block, arrows highlighting motion artifacts).
  • Adaptive Multi-contrast MR Image Denoising based on a Residual U-Net using Noise Level Map
    Jiahao Hu1,2,3, Yilong Liu1,2, Zheyuan Yi1,2,3, Yujiao Zhao1,2, Fei Chen3, and Ed X. Wu1,2
    1Laboratory of Biomedical Imaging and Signal Processing, The University of Hong Kong, Hong Kong, China, 2Department of Electrical and Electronic Engineering, The University of Hong Kong, Hong Kong, China, 3Department of Electrical and Electronic Engineering, Southern University of Science and Technology, Shenzhen, China
    This study presents an adaptive multi-contrast MR image denoising based on a residual U-Net using a noise level map. The introduced noise level map can be manually set to fit different noise levels. The denoising results outperform BM3D in noise reduction and details preservation.
    Fig. 1. (a) The architecture of the proposed multi-contrast denoising method using residual U-Net by combining U-Net and ResNet. (b) Residual blocks consisting of two convolutional layers with a ReLU activation in between. (c) Strided conv2D block consisting of a convolutional layer and ReLU activation. (d) Transposed conv2D block consisting of a transposed convolutional layer and ReLU activation.
    Fig. 3. The denoising results for images with a higher noise level. The same level of noise (σ=25) was added to form noisy images. The proposed method remained similar performance, while BM3D smoothed the image details even more if attempt to reduce the noise to the same level as the proposed method.
  • Simultaneous morphological and quantitative lumbar MRI with 3D isotropic high-resolution using MIXTURE T2
    Daichi Murayama1, Takayuki sakai1, Masami Yoneyama2, and Shigehiro Ochi1
    1Radiology, Eastern Chiba Medical Center, Chiba, Japan, 2Philips Japan, Tokyo, Japan
    MIXTURE could simultaneously provide morphology, pathology and T2 quantitative lumbar imaging in one single scan. It's useful to  quantitatively evaluate the tissues around lumbar for 3D T2 mapping and improve the ability of diagnostic imaging around the lumbar for 3D multi contrast MRI.

    Fig.1 Scheme of the MIXTURE (Multi-Interleaved X-prepared tse with inTUitive RElaxometry).

    (a) T2-mapping was performed using T2-prepared 3D segmented turbo spin- echo (TSE) with variable refocusing pulse trains. (b) Two images with different TE (TE = 0 and 50ms) were acquired with interleaved acquisition. To obtain the compatible contrasts with routine TSE images, TSE shot#1 did not apply any pre-pulses (as “PDW) and shot#2 applied both SPAIR and T2pre (as “fat-suppressed T2W”).

    Fig.4 Clinical images with low back pain using MIXTURE.

    (A) MIXTURE, Multiple Interleaved X-prepared TSE with Universal Relaxation-mapping Extensions,(B) Conventional 2D TSE

  • Accelerated MR parametric mapping with a hybrid deep learning model
    Haoxiang Li1,2, Jing Chen1, Yuanyuan Liu1, Hairong Zheng1, Dong Liang1, and Yanjie Zhu1
    1Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China, 2Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Shenzhen, China
     This work proposes a deep learning based method to accelerate MR parametric mapping both by reducing the contrast number and undersampling the k-space data.
    Fig. 1 (a) The proposed MRI parameter mapping method consists of two neural networks module and a pixel-wise curve fitting process. (b) The reconstruction module is based on deep ADMM-Net for reconstruction of weighted images from under-sampled k-space data. (c) The generative module is a densely connected neural network to generate the corresponding weighted images from the reconstructed image pairs under the supervision of corresponding weighted images.
    Fig. 5 The ROI analysis of test $$$T_{1\rho}$$$ knee data (R=1) and error map of test $$$T_{1\rho}$$$ knee data (R=1); Proposed: Mapping with 2 acquired fully sampled images and 3 synthetic images (Equivalent R = 2.5); Two contrast images: Mapping with 2 acquired fully sampled images only (Equivalent R = 2.5).
  • Simultaneous 3D T1 weighted, T2 weighted and FLAIR imaging using Serial Connection of Echo Train Acquisitions (SCETA) for Multi-contrast imaging
    Naoyuki Takei1, Shohei Fujita2,3, Issei Fukunaga2, Mitsuharu Miyoshi1, Shigeki Aoki2, Suchandrima Banerjee4, and Tetsuya Wakayama1
    1MR Applications and Workflow, GE Heatlcare, Tokyo, Japan, 2Juntendo University School of Medicine, Tokyo, Japan, 3The University of Tokyo Graduate School of Medicine, Tokyo, Japan, 4MR Applications and Workflow, GE Heatlcare, Menlo Park, CA, United States
    A novel 3D multi-contrast imaging technique using the hybrid acquisition of FSE and GRE provided T1 weighted, T2 weighted and FLAIR images simultaneously. The comparison with conventional 3D imagings in term of image contrast and scan time demonstrated a promising result.
    Fig.1. (a) A schematic pulse sequence diagram of SCETA (Serial Connection of Echo Train acquisition) to acquire T1w, T2w and FLAIR using the hybrid acquisition of FSE and GRE. (b) Echo signal of white matter, gray matter and CSF builds three imaging contrasts in a TR.
    Fig. 2. A comparsion result with conventional 3D scan of T2w, FLAIR and T1w. SCETA reduces scan time by a factor of approximately 2.4 while maintaining similar image appearance to conventional scan.
  • Rapid and Simultaneous Acquisition of T2-weighted and Fluid-attenuated Brain Images using a Spiral-ring Turbo Spin-echo Imaging
    Zhixing Wang1, Xue Feng1, John Mugler2, and Craig Meyer1
    1Biomedical Engineering, University of Virginia, Charlottesville, VA, United States, 2Radiology & Medical Imaging, University of Virginia, Charlottesville, VA, United States
    This study describes a new approach to obtain T2-weighted and fluid-attenuated inversion recovery (FLAIR) images simultaneously in a short scan time using a spiral-ring Turbo Spin-echo sampling strategy combined with a 180°(y)-90°(x) driven-inversion preparation RF pulse.
    Figure 1. Pulse sequence diagram showing the sampling strategy, which includes the time-multiplexed multislice scheme, SPRING TSE in/out variant data acquisition, and driven-inversion RF pulses. The boxes with stripes show slices for the T2-weighted acquisition, while the open boxes show slices for the FLAIR acquisition. The time interval between boxes with the same number is set to TI. A SPRING TSE in/out variant sampling scheme is used in each box for data acquisition, while a 180°-90° RF pair is applied at the end of the echo train for boxes with stripes to achieve driven inversion.
    Figure 3. Comparison of in-vivo images acquired using standard Cartesian TSE for T2-weighted images, Cartesian FLAIR for fluid-attenuated images, and the proposed method for both the T2-weighted and fluid-attenuated images. Both the T2-weighted images (top panel) and FLAIR images (bottom panel) from the proposed method show image quality and contrast similar to the conventional images.
  • Merging T1 weighted images with QSM provides a unique contrast for brain tissue segmentation in humans and non-human primates
    Rakshit Dadarwal1,2 and Susann Boretius1,2
    1Functional Imaging Laboratory, German Primate Center, Göttingen, Germany, 2Georg August Universität Göttingen, Göttingen, Germany
    We have proposed a method to combine T1w and QSM contrasts that significantly improves the quality of subcortical nuclei classification while retaining the excellent white matter delineation of T1w.
    Figure 1. The juxtaposition of a human subject’s T1–QSM fusion image (TQ-SILiCON map) in comparison with the respective T1 weighted image (A, left) and QSM (B, left). TQ-SILiCON maps were generated with the weights of 0.318 and -0.790 for T1w and QSM respectively. 1 – thalamus, 2 – external globus pallidus, 3 – internal globus pallidus, 4 –substantia nigra, 5 – red nucleus, 6 – white matter, 7 – internal capsule, and 8 – pulvinar nucleus.
    Figure 2. Illustration of a macaque’s T1-QSM fusion image (TQ-SILiCON map) in comparison with the respective T1 weighted image (A, left) and QSM (B, left). The emphasized areas on the TQ-SILiCON map are parts of the subcortical gray matter nuclei: 1 – thalamus, 2 – external globus pallidus, 3 – internal globus pallidus, 4 – substantia nigra, 5 – red nucleus, and 6 – white matter.
  • Comprehensive multiparametric cardiac MRI tissue phenotyping (LGE, T1, T2, DWI, BOLD & VAI) of acute myocardial infarction in swine
    Holly Doig1, Maaike van den Boomen1,2,3, Erin Connors1, Joan Kim1, Jaume Coll-Font1,3, Robert A. Eder1, Shi Chen1, Yoshiko Iwamoto1, Kyrre E. Emblem4, Kawin Setsompop3, Niek H.J. Prakken2, Ronald J.H. Borra2,5, and Christopher T. Nguyen1,3
    1Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, United States, 2Department of Radiology, University Medical Center Groningen, Groningen, Netherlands, 3A.A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, United States, 4Department of Diagnostic Physics, Oslo University Hospital, Oslo, Norway, 5Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, Groningen, Netherlands
    Comprehensive multiparametric cardiac MRI tissue phenotyping (LGE, T1, T2, DWI, BOLD and VAI) of acute myocardial infarction in swine show that only LGE, T2, and BOLD consistently detect acute infarct. Both T1 and DWI were moderately elevated but not significant across all groups.
    Figure 1 - Overall study design of inducing myocardial infarction in Yorkshire pigs and imaging 3 days post MI before sacrificing animal to perform histological validaiton.
    Figure 3 – Polar AHA plots of example animal with corresponding segment based bar graphs. With a visible increased segment 2 and 3 (2036±132 [a.u]) in the LGE plot indicating the infarct and segment 5 as remote segment (SI of 1844±48 [a.u.]). The T1 in those segments was non-significantly increase to 1260±56ms but the remote segment also showed a high T1 value of 1275±42ms. The infarct T2 was 48±4ms, which was a significant increase compared to the remote 35±2ms. The ADC values was also increased 1933±95.25x10-6mm2/s compared to 1839±123x10-6mm2/s in the remote, but was not significant.
  • Stroke analysis with fully automatic multi-contrast MR image registration
    Weijian Huang1, Yulon Qi2, Qiang He3, Ting Ma4, Xin Liu1, Guanxun Cheng2, Hairong Zheng1, and Shanshan Wang1
    1Paul C Lauterbur Research Center, Shenzhen Institutes of Advanced Technology Chinese Academy of Sciences, Shenzhen, China, 2Radiology department, Peking University Shenzhen Hospital, shenzhen, China, 3United Imaging Research Institute of Innovative Medical Equipment, Shenzhen, China, 4Pengcheng Laboratory, Shenzhen, China
    Identifying stroke lesions is crucial in clinic. The misalignment between multi-contrast images is the main obstacle in this process. This paper proposes a new registration framework to for accurate and quick multi-contrast image registration, with promising performances achieved. 
    Fig. 3. Qualitative results of different methods. The blue and red edge lines indicate the stroke lesion regions annotated by radiologists based on DWI and FLAIR, and the green lines indicate the predictions of different methods.
    Fig.2. Flow chart of the proposed method. M represents the moving image, F represents the fixed image, and M-1D represents the prediction of the inverse transformation. We impose MSE constraints on M-1D and MA to ensure topological deformation.
  • In-vivo ferumoxytol imaging and T1/T2 characterization at 64mT
    Thomas Campbell Arnold1, Samantha By2, Hadrien Dyvorne2, Rafael O'Halloran2, Farzana Sayani3, Lisa M. Desiderio4, Brian Litt1,5, and Joel M. Stein4
    1Bioengineering, University of Pennsylvania, Philadelphia, PA, United States, 2Hyperfine Research, Guilford, CT, United States, 3Medicine, Perelman School of Medicine, Philadelphia, PA, United States, 4Radiology, Perelman School of Medicine, Philadelphia, PA, United States, 5Neurology, Perelman School of Medicine, Philadelphia, PA, United States
    Ferumoxytol is a promising contrast agent in low-field MRI. We characterize ferumoxytol T1/T2 relaxation properties and collect in-vivo imaging of iron-deficiency anemia patients. We observed contrast enhancement on T1/T2/FLAIR imaging and venous/arterial contrast in an angio sequence.
    T1-weighted angiographic imaging with ferumoxytol at 64mT showing enhancement of dural venous sinuses and jugular veins, distal cervical and intracranial internal carotid arteries (ICA) as well as middle cerebral arteries (MCA, arrows).
    Contrast enhancement with ferumoxytol on standard sequences optimized for brain tissue contrasts at 64 mT. On T1 images, ferumoxutol modestly increases signal in the superior sagittal sinus (arrows) and other venous structures. On T2 and T2-FLAIR images ferumoxytol markedly decreases signal of intrinsically hyperintense veins, shown as enhancement on inverse difference images (i.e. pre-minus-post).
Back to Top
Digital Poster Session - Novel Contrast Mechanisms
Contrast Mechanisms
Monday, 17 May 2021 15:00 - 16:00
  • BOLD-free fMRI?
    Tobias C Wood1 and Nikou Louise Damestani1
    1Neuroimaging, King's College London, London, United Kingdom
    We demonstrate a proof-of-concept fMRI experiment using a BOLD-insensitive MT-prepared ZTE pulse sequence, which exploits the recently proposed Arterial Blood Contrast mechanism. We show tightly localised responses to a visual checkerboard task in a small number of subjects.
    Figure 4: Results of the single-subject analyses overlaid in MNI space. At both 480° and 500° power levels focal activation in the visual cortex can be observed. At 0° MT power, apparent activation can be observed in the sagittal sinus, most likely due to blood volume changes within the sinus.
    Figure 3: Temporal SNR achieved for the three different power levels. The reduction of signal due to MT leads to a notable fall in tSNR.
  • High neuromelanin contrast achieved using sandwiched flow saturation RF pulses: sandwich-fsNM imaging
    Sooyeon Ji1, Eun-Jung Choi1, Eung Yeop Kim2, Dong Hoon Shin3, Hyeong-Geol Shin1, and Jongho Lee1
    1Department of Electrical and Computer Engineering, Seoul National University, Seoul, Korea, Republic of, 2Department of Radiology, Gachon University Gil Medical Center, Incheon, Korea, Republic of, 3Department of Neurology, Gachon University Gil Medical Center, Incheon, Korea, Republic of
    A 3D neuromelanin-sensitive imaging protocol is developed using a GRE sequence with multiple flow saturation pulses for magnetization transfer weighting. The proposed protocol displays higher neuromelanin contrast compared to conventional methods.
    Figure 5. Comparison of the images acquired with the proposed protocol, MT-GRE, and MT-TSE. When calculated for the ROIs in b), the contrast of the sandwich-fsNM is superior to both MT-GRE and MT-TSE imaging in terms of CR and CNR. SNR of sandwich-fsNM is lower than MT-GRE but higher than MT-TSE. MT-GRE images suffer from flow artifacts.
    Figure 4. Simulated and experimental CR between SN and CC with respect to a) Dflowsat and b) number of FSs (Nflowsat). The CR increases as Dflowsat decreases and as Nflowsat increases, both in simulations and experiments. The effect size of decreasing Dflowsat is relatively smaller than that of increasing Nflowsat. c) In the experimental data, CNR shows similar trends to CR whereas SNR decreases as Dflowsat decreases and as d) Nflowsat increases.
  • EPR measurements on human brain tissue at variable temperature
    Fábio Seiji Otsuka1, Carlos Salmon1, Otaciro Nasimento2, and Maria Otaduy3
    1Physics Department, University of São Paulo, Ribeirão Preto, Brazil, 2Physics Institute, University of São Paulo, São Carlos, Brazil, 3Medical School, University of São Paulo, São Paulo, Brazil

    Four different peaks were observed on all brain regions. The rhombic iron, copper and organic radical peaks presented a Curie behavior while the broad peak at g = 2.0 presented an antiferromagnetic behavior. Finally, the Locus Coeruleus showed an additional peak.

    Figure 1 – (a-e) EPR spectra and the fitting of GP sample at different temperatures. Inserts represent the spectrum region around g = 2.0. f) The organic radical peak after subtraction of other fitted peaks.
    Figure 2 – Second integral and linewidth as a function of temperature for each peak (Fe_hs, Cu, broad peak at g=2.0 and the organic radical) and each sample (SN, GP, LC). For the Fe_hs, Cu and organic radical peaks fitting was performed using Curie-Weiss Law. For the broad peak Vav-Vleck paramagnetism for S=5/2 and S=1/2 with Curie-Weiss law was used for fitting.
  • Radiofrequency heating measurement using MR thermometry and field monitoring: methodological considerations and first in vivo results.
    Caroline Le Ster1, Franck Mauconduit1, Christian Mirkes2, Michel Bottlaender3,4, Fawzi Boumezbeur1, Boucif Djemai1, Alexandre Vignaud1, and Nicolas Boulant1
    1Paris-Saclay University, CEA, CNRS, BAOBAB, Neurospin, Gif-sur-Yvette, France, 2Skope MRT, Zurich, Switzerland, 3Paris-Saclay University, CEA, CNRS, INSERM, BioMaps, Service hospitalier Joliot, Orsay, France, 4UNIACT, Neurospin, CEA, Gif-sur-Yvette, France
    Inclusion of field fluctuations in MR thermometry experiments performed at 7T with concurrent radiofrequency heating on an agar-gel phantom and on an anaesthetized macaque showed beneficial for the measurement of small temperature rises as encountered in standard brain exams. 
    Figure 2: Scan without RF heating results for the macaque (run 1). (a) Mean ΔT maps and (b) corresponding SD maps computed from voxel-wise detrended signals. ΔT maps were computed from images reconstructed with nominal trajectories (upper row) and first order spherical harmonics field correction (lower row). ΔT computed from phase images corrected with rotation regressors around LR, AP and HF axis are also displayed. (c,d) Rotation parameters and (e,f) time evolution of ΔT computed from nominal and FS-corrected images, respectively, in the voxel indicated by the white cross in (a).
    Figure 1: (a) Temperature variation measured in vitro with the fiber optical probe and from nominal and field sensor (FS) corrected MRT images at probe location over the 20 minutes scan at approximately 0% of the maximal SAR. (b) Temperature variation measured during the heating scan (5 min pre-heating at 0% SAR + 20 min heating at maximal SAR + 5 min post-heating at 0% SAR) by the fiber optical probe and from nominal and FS-corrected MRT images at probe location, in field disturbed conditions.
  • Experimental Evaluation of Spin Echo based Magnetic Resonance Magnetohydrodynamic Flow  Velocimetry
    Mert Şişman1, Mehdi Sadighi1, Hasan Hüseyin Eroğlu2,3, and B. Murat Eyüboğlu1
    1Electrical and Electronics Engineering, Middle East Technical University (METU), Ankara, Turkey, 2Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital, Amager and Hvidovre, Denmark, 3Center for Magnetic Resonance, DTU Health Tech, Technical University of Denmark, Kgs Lyngby, Denmark
    Magnetohydrodynamic (MHD) flow occurs due to the Lorentz force formed by the interaction between the static magnetic field of the MR scanner and the externally injected electric current. In this study, simulated and  experimental MHD flow velocity distributions are obtained and validated.
    Figure 5: MHD flow velocity distributions of the homogeneous experimental phantom: x-component with current injection parameters (a) I = 10 mA, TC = 10 ms, (b) I = 5 mA, TC = 10 ms, and (c) I = 10 mA, TC = 5 ms; y-component with current injection parameters (d) I = 10 mA, TC = 10 ms, (e) I = 5 mA, TC = 10 ms, and (f) I = 10 mA, TC = 5 ms; and z-component with current injection parameters (g) I = 10 mA, TC = 10 ms, (h) I = 5 mA, TC = 10 ms, and (i) I = 10 mA, TC = 5 ms.
    Figure 4: MHD flow velocity distributions of the homogeneous simulation model: x-component with current injection parameters (a) I = 10 mA, TC = 10 ms, (b) I = 5 mA, TC = 10 ms, and (c) I = 10 mA, TC = 5 ms; y-component with current injection parameters (d) I = 10 mA, TC = 10 ms, (e) I = 5 mA, TC = 10 ms, and (f) I = 10 mA, TC = 5 ms; and z-component with current injection parameters (g) I = 10 mA, TC = 10 ms, (h) I = 5 mA, TC = 10 ms, and (i) I = 10 mA, TC = 5 ms.
  • Simultaneous Magnetic Resonance Magnetohydrodynamic Flow Velocity and Diffusion Tensor Imaging
    Mert Şişman1, Mehdi Sadighi1, and B. Murat Eyüboğlu1
    1Electrical and Electronics Engineering, Middle East Technical University (METU), Ankara, Turkey
    In this study, it is shown that the magnetohydrodynamic (MHD) flow velocity and diffusion tensor images can be obtained from the same acquisition with careful selection of the flow-encoding gradients. The experimental results demonstrate validity of this claim.
    Figure 3: MHD flow velocity distributions of the experimental phantom with vertical current injection. Using the conventional flow-encoding set ($$$\mathbf{{G_f^k}}$$$): (a) vx, (b) vy, and (c) vz; and using the proposed flow-encoding set ($$$\mathbf{{G_f^k}^*}$$$): (d) vx, (e) vy, and (f) vz. The $$$RMSE(\%)$$$ values in each direction: $$$RMSE_x=2.17\%$$$, $$$RMSE_y=2.80\%$$$, and $$$RMSE_z=1.66\%$$$.
    Figure 5: Diffusion tensor images obtained from simultaneous DTI and MHD imaging. $$$\overline{\overline{D}}$$$ distributions obtained without current injection: (a) dxx, (b) dyy, and (c) dzz; with vertical current injection: (d) dxx, (e) dyy, and (f) dzz; and with horizontal current injection: (g) dxx, (h) dyy, and (i) dzz. $$$\overline{\overline{D}}$$$ images obtained with current injections have $$$\sqrt2$$$ SNR advantage.
  • Theoretical evaluation of the feasibility to detect label-exchange by proton MRS at 7T in human brain after administration of deuterated glucose.
    Simone Poli1, Lia Bally2, Roland Wiest3, and Roland Kreis1
    1Department of Radiology and Biomedical Research, University of Bern, Bern, Switzerland, 2Department of Diabetes, Endocrinology, Nutritional Medicine and Metabolism, Inselspital, Bern University Hospital and University of Bern, Bern, CH, Bern, Switzerland, 3Institute of Diagnostic and Interventional Neuroradiology, Inselspital, Bern University Hospital and University of Bern, Bern, CH, Bern, Switzerland
    To evaluate feasibility of indirect 1H MRS detection of metabolic changes after intake of deuterated glucose, brain spectra were simulated and minimal fitting uncertainties determined. We expect labeling of glutamate, lactate and glutamine to be observable, for large VOIs, but also in MRSI
    Figure 1. a) Simulated basis spectra with expected in vivo lineshape at equal concentrations for glutamate, glutamine, lactate, glucose, and their mono- and double- deuterated counterparts. b) Synthetic normal brain spectrum with main metabolite peaks labeled vs. the expected brain spectrum after supplementation of deuterated glucose with effect size taken from Ref. 3 and c) difference between the two, amplified by a factor of 15 for better visualization.
    Figure 2. Comparison of simulated and measured phantom spectra of [6,6’-2H2]-Glc and [6,6’-1H2]-Glc at high resolution to verify the appropriateness of the simulations and thus the applicability of the precision estimation based on CRB calculations in synthetic spectra with in vivo conditions.
  • Relaxation Anisotropy in Biological Tissues
    Nina Elina Hänninen1,2, Mikko Johannes Nissi1,2, Matti Hanni1,3,4, Olli Gröhn5, Miika Tapio Nieminen1,3,4, and Timo Liimatainen1,4
    1Research Unit of Medical Imaging, Physics and Technology, University of Oulu, Oulu, Finland, 2Department of Applied Physics, University of Eastern Finland, Kuopio, Finland, 3Medical Research Center, University of Oulu and Oulu University Hospital, Oulu, Finland, 4Department of Diagnostic Radiology, Oulu University Hospital, Oulu, Finland, 5A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
    Cartilage and tendon represented highest relaxation anisotropy in comparison to brain, spinal cord, kidney and cardiac muscle tissue. T2 presented highest anisotropy, while rotating frame relaxations exhibited less anisotropy, and T1 showed no anisotropy.
    Figure 1. T2-weighted images (left), T2 maps in 5 different orientations with respect to B0 (middle), and calculated T2 relaxation anisotropy maps (right) for representative tissue samples: A) Articular cartilage (bovine patella); B) Tendon (bovine ACL); C) Brain tissue (mouse, right hemisphere, coronal view); D) Spinal cord and muscle (mouse, sagittal view); E) Cardiac muscle (mouse, coronal view); F) Kidney (mouse, coronal view). The color scales are adjusted separately for each sample.
    Figure 2. Relaxation anisotropy maps of different parameters, T1, CW-T1ρ (500 Hz), adiabatic T1ρ, RAFF2, and T2 in representative tissue samples: A) Articular cartilage (bovine patella); B) Tendon (bovine ACL); C) Brain tissue (mouse, right hemisphere, coronal view); D) Spinal cord and muscle (mouse, sagittal view); E) Cardiac muscle (mouse, coronal view); F) Kidney (mouse, coronal view). The color scales are adjusted separately for each sample.
  • Towards high precision thermal based RF safety assessment with cardiac triggered MR thermometry
    Bart R. Steensma1, Cornelis A.T. van den Berg1, and Alexander J.E. Raaijmakers2
    1Center for Image Sciences - Computational Imaging Group, University Medical Center Utrecht, Utrecht, Netherlands, 2Biomedical Engineering - Medical Imaging Analysis, Eindhoven University of Technology, Eindhoven, Netherlands
    By using a PPU during MR Thermometry, we were able to perform reproducible thermometry measurements of in vivo local temperature rise of less than 1 °C 
    Figure 4: 4a. spatial maps of temperature rise calculated from MRT measurements after drift correction. The images in the top row were acquired with no RF heating, while the images on the two bottom rows were acquired with the same RF shim. 4b. Time series of temperature change in the indicated voxels for all 3 measurements.
    Figure 3: 3a and 3b. Spatial maps of temporal standard temperature deviation the upper leg during a MRT scan where no RF heating is applied, without and with cardiac triggering. 3c and 3d. Time series of temperature deviations in indicated voxels (3a and 3b). No drift correction was applied yet during this short time interval.
  • Virtual non-contrast enhanced MRI (VNC-MRI)
    Thomas Lindner1, Hanna Debus1, and Jens Fiehler1
    1University Hospital Hamburg-Eppendorf, Hamburg, Germany
     This study presents an approach to retrospectively remove contrast agent from the final images, denoted as "virtual non-contrast enhanced imaging". Virtual non-contrast imaging can be performed by acquiring images with different flip angles and characterizing the individual signals.
    Figure 4: In-vivo scans of patient one. Image acquired with a low flip angle of 5° (A) and the corresponding VNC image (B). Image acquired with a high flip angle of 26° (C) and the corresponding VNC image (D). T1weighted image acquired after CA injection (E), true non-contrast enhanced image (F).
    Figure 5: In-vivo scans of patient two. Image acquired with a low flip angle of 5° (A) and the corresponding VNC image (B). Image acquired with a high flip angle of 26° (C) and the corresponding VNC image (D). T1weighted image acquired after CA injection (E), true non-contrast enhanced image (F).
  • Monte Carlo Simulation for Magnetic Nanoparticle Biosensors
    Tristhal Parasram1, Rongsheng Lu2, Yi Chen2, and Dan Xiao1
    1University of Windsor, Windsor, ON, Canada, 2Southeast University, Nanjing, China
    A Monte Carlo simulation is developed to investigate the effectiveness of magnetic nanoparticle biosensors. This simulation will be employed to optimize the nanoparticle biosensor systems for a wide range of targets, including cancer cells and COVID virus.
    Simulated magnetic field distribution (in Hz) and CPMG signals for (a) grid, (b) random, and (c) clustered nanoparticle configurations, with the same number of particles. The apparent T2 values were 0.2, 0.4 and 0.7s for (a), (b) and (c), respectively.
    T2 relaxation times of different concentration iron nanoparticles in water. The red points are simulation results and the solid black points are experimentally acquired. The simulated bulk T2 of pure water was 2 s. 128 CPMG data points were acquired with 32000 diffusing water molecules.
  • Feasibility of Magnetic Resonance Thermometry at 0.55T
    Waqas Majeed1, Axel J. Krafft2, Sunil Patil1, Henrik Odéen3, John Roberts3, Florian Maier2, Dennis L. Parker3, and Himanshu Bhat1
    1Siemens Medical Solutions USA Inc., Malvern, PA, United States, 2Siemens Healthcare GmbH, Erlangen, Germany, 3Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT, United States
    We demonstrate that high quality PRF thermometry can be achieved in the brain and prostate at 0.55T using a segmented EPI approach. 
    Figure 3: σT in the brain with ETL 49, TE 81, 3.8s/frame: σT values of less than 2.5°C are overlaid on average magnitude image (8 out of 12 sagittal slices). σT is less than 1°C in most of the brain, demonstrating excellent precision. Average σT of 0.7 ± 0.2°C was observed in the ROI indicated on the figure. This protocol offers a higher frame rate compared with that with ETL 33. The slight reduction in σT, as compared with the protocol with ETL 33, can be attributed to the longer TE.
    Figure 5: σT in the prostate with TR 100ms, TE 45ms, 7.2 s/frame. σT values of less than 2.5°C are overlaid on average magnitude image (8 out of 12 axial slices; cropped to show details). Average σT of 1.4±0.3°C was observed in the ROI indicated on the figure, demonstrating good precision.
  • Improved PRF-based MR Thermometry for Tissues with Aqueous and Adipose
    Chang-Sheng Mei1, Shenyan Zong2, and Guofeng Shen2
    1Department of Physics, Soochow University, Taipei, Taiwan, 2Biomedical Instrument Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
    In this work, the conventional PRF method was evolved using the circle fitting for the temperature measurements in fat-containing tissues. As a result, the temperature accuracy by the proposed method in aqueous and adipose tissues was improved in our verification experiments.
    Fig. 1: The diagrams of water and fat phasors of MR data in Cartesian coordinates. In (a), the add-up phasor represents the actual phasor obtained from image, where the orange and blue arrows are phasors for fat and water protons, respectively. (b,c) show the water phase rotation around point 'O' caused by temperature changes and depict two scenarios of underestimating (φ2 < φ1) and overestimating (φ2 > φ1), respectively. A circle fitting can be applied here to determine the circle center 'O', which represents the fat phasor AO in (b) and (c), for getting the accurate water phase changes.
    Fig. 3: Results of in vitro phantom experiments. In (a), the red pentagon and filled circle indicate the phantom region and the selected voxel for the plots of temperature evolution over time in (b). The orange, blue and red lines display the temperature changes obtained by the conventional method, the proposed method and the optical fiber. Approximately 33% of temperature error caused by 15% fat in phantom was observed and corrected in (b).
  • Effects of T2* on accuracy of single reference variable flip angle T1 – mapping for MR thermometry
    Michael Malmberg1, Dennis L Parker2, and Henrik L Odéen2
    1Biomedical Engineering, University of Utah, Salt Lake City, UT, United States, 2Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT, United States
    Neglecting T2* changes in the single reference variable flip angle (SR-VFA) method for T1-mapping produces a systematic bias on T1. This bias can be corrected by measuring T2* changes dynamically, at the expense of noise, which noise could be mitigated through weighted T1-map combinations.
    Figure 2: T1 calculation bias for standard VFA method, SR-VFA method, and T2* corrected SR-VFA method. TR = 10 ms, T1 baseline = 350 ms, TE/T2* = 0.2;. SR-VFA line is equivalent with the bolded lines in Figure 1.
    Figure 1: (left) – T1 calculation bias vs Z ratio with TE/T2* = 0.2; (right) – T1 calculation bias vs TE/T2* with Z ratio = 0.5; For both plots – TR = 10 ms, T1 baseline = 350 ms, α = 6.4°, β = 21.2°. Bolded lines are equivalent.
  • Accuracy of MR thermometry during deep radiofrequency hyperthermia treatments in the pelvic region
    Iva VilasBoas-Ribeiro1, Sergio Curto1, Gerard C. van Rhoon1,2, and Margarethus M. Paulides1,3
    1Department of Radiotherapy, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, Netherlands, 2Department of Radiation Science and Technology, Faculty of Applied Sciences, Delft University of Technology, Delft, Netherlands, 3Center for Care and Cure Technologies Eindhoven (C3Te), Department of Electrical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
    This study indicates that change in gastrointestinal air volume can be used as a criteria for selection of patients with higher accuracy in MR thermometry. We showed that this change can be exploited for imaging based selection criteria prior to hyperthermia treatment.
    Figure 5: Quantification of MR thermometry accuracy in all patients and in the selected patients. The red dotted line represents the thresholds for acceptance.
    Figure 3: Temperature distributions acquired during treatment from the representative patient, where the time corresponds to the minutes from the time that the RF power was applied. The slice presented correspond to the middle slice (yellow line from Figure 1).
  • Towards correlating tissue status with dynamic PRF-T1 measurements using a single reference dual flip angle technique
    Henrik Odéen1, Sara L Johnson1, Allison H Payne1, and Dennis L Parker1
    1Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT, United States
    A single-reference dual flip angle method is used for simultaneous volumetric PRF/T1 measurements in a clinically relevant time. The slope of the T1 vs. temperature curve correlates with delivered thermal dose. T1 can be used as a complimentary measure for treatment outcome evaluation.
    Figure 4. T1 (ms) as a function of temperature (°C) for the 14 consecutive sonications in experiment 2. Mean +/- standard error over 3x3 voxels is shown for heating (red) and cooling (blue), and lines indicate linear fits. The title for each sub-plot states the slope of the heating and cooling part, respectively. The last 4 sonications delivered increasing amounts of dose, from 41 to 1*108 CEM43.
    Figure 3. Temperature (°C) and T1 (ms) as a function of time for experiment 2. The first five sonications used 29 eW, the next five used 52 eW, and the last four used 66 eW, all for 19.6 s with 39.2 s cooling in-between consecutive sonications.
  • Analysis of neurodegeneration using diffusion and functional MRI in FTLD model marmoset
    Mitsuki Rikitake1, Junichi Hata2, Fumiko Seki3, Shinsuke Ishigaki4, Kuniyuki Iwata-Endo4, Nobuyuki Iwade4, Takako Shirakawa1, Hirotaka James Okano2, Hideyuki Okano5, and Gen Sobue4
    1Department of Redioligical Science, Human Health Science, Tokyo Metroplitan University, Tokyo, Japan, 2Jikei University Graduate School of Medicine, Tokyo, Japan, 3Central Institute for Experimental Animals, Kanagawa, Japan, 4Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan, 5RIKEN Center of Brain Science, Saitama, Japan
    We analyzed the degenerate in cranial nerve structure and change brain function in the marmoset FTLD model. Image analysis using structural MRI detected gray matter degeneration.Further, fMRI detected decrease in activity of the brain region, which was correlated with brain degeneration.
    The results of image analysis of VBM and VBA in GM are shown.The cross-sectional image is a coronal image.In VBA analysis, from left to right, AD/FA/RD in DTI analysis, and ODI/FICVF in NODDI analysis are presented.For each image parameter, if group injected with FUS is significantly higher(p<0.05) with respect to group injected with saline (upper) before injection FUS (lower), it is represented as red, and if group injected with FUS is significantly lower(p<0.05), it is represented as blue.
    The results of image analysis of VBM and VBA in WM are shown.The cross-sectional image is a coronal image.In VBA analysis, from left to right, AD/FA/RD in DTI analysis, and ODI/FICVF in NODDI analysis are presented.For each image parameter, if group injected with FUS is significantly higher(p<0.05) with respect to group injected with saline (upper) before injection FUS (lower), it is represented as red, and if group injected with FUS is significantly lower(p<0.05), it is represented as blue.
  • The Use of the 3He/129Xe MRI Lung Morphometry for a Longitudinal Observation of the Emphysema Progression in AATD Patients
    Elise Noelle Woodward1, Matthew S Fox1,2, Tingting Wu3, Hacene Serrai1, David G McCormack4, Grace Parraga3,4,5,6, and Alexei Ouriadov1,2,6
    1Physics and Astronomy, Western University, London, ON, Canada, 2Lawson Health Research Centre, London, ON, Canada, 3Department of Medical Biophysics, Western University, London, ON, Canada, 4Department of Medicine, Respirology, Western University, London, ON, Canada, 5Robarts Research Institute, London, ON, Canada, 6School of Biomedical Engineering, Western University, London, ON, Canada
    We demonstrated that normalizing inconsistencies in ADC/Lm based emphysema progression biomarkers is an important step in increasing the validity of such measurements. Ventilation defect percentage is a possible way to remedy these inconsistencies.
    AATD=Alpha-one antitrypsin deficiency; AATD-1= Ex-smoker AATD; ADC = apparent diffusion coefficient; VDP = ventilation defect percentage; Lm = mean linear intercept estimate.
    Figure 4 shows five representative ADCHe/ADCXe maps for the AATD-1 subject.
  • Metabolite-specific echo planar imaging of hyperpolarized 13C-pyruvate at 4.7T
    Tyler Blazey1, Galen D Reed2, Joel R Garbow1, and Cornelius von Morze1
    1Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, United States, 2GE Healthcare, Dallas, TX, United States
    We developed a metabolite-specific 2D EPI sequence with spatially-selective RF pulses for [1-13C]pyruvate and [1-13C]lactate at 4.7T. Using this sequence for in vivo imaging of hyperpolarized [1-13C]pyruvate resulted in improvements in temporal and spatial resolution compared to CSI.
    Figure 1: Pulse sequence diagram for metabolite-specific 2D EPI imaging of [1-13C]pyruvate and [1-13C]lactate at 4.7T. Lactate-only excitation pulse is shown.
    Figure 3: Metabolite images of [1-13C]pyruvate (1st column) and [1-13C]lactate (2nd column) in the liver and kidney of a rat following injection of [1-13C]pyruvate. Images in the first row were acquired using metabolite-specific EPI (Figure 1) while images in the second row were acquired using a standard CSI sequence. All images were obtained by summing signal over all time points. White lines are acquisition grids for each image. Images are shown with a 2x interpolation factor.
  • Feasibility of model-based omega-3 fatty acid fraction mapping using multi-echo gradient-echo imaging at 3T
    Dominik Weidlich1, Julius Honecker2, Claudine Seeliger2, Daniela Junker1, Marcus R. Makowski1, Hans Hauner2, Dimitrios C. Karampinos1, and Stefan Ruschke1
    1Department of Diagnostic and Interventional Radiology, School of Medicine, Technical University of Munich, Munich, Germany, 2Else Kröner Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, Germany
    First feasibility of imaging-based ω-3 fraction mapping was demonstrated in phantoms, as well as in vitro and in vivo in human adipose tissue. In the phantom experiment, promising correlations were obtained between imaging-based parameters and chromatography–mass spectrometry.
    Figure 5: Resulting parameter maps of the thigh from the in vivo experiment: a) ω-3 fatty acid fraction, b) PDFF, c) T2*, d) ndb, e) nmidb and f) CL. A ROI analysis (see ROI #1 in b)) yielded ω-3: 25.4±7.5%, ndb: 2.41±0.40, nmidb: 0.67±0.26, CL: 18.41±0.54, T2*: 30.7±6.9ms. Low PDFF regions (e.g. muscle tissue) were masked out in a) and e)-f).
    Figure 2: Obtained parameter maps from the phantom experiment and correlation plots for the ω-3 fraction, ndb, nmidb and CL. ROI placement is indicated in red color in b) the PDFF map. Fig.3 shows the correlation of the ROI measurements with GC-MS calibrated reference values.