Imaging Metabolites: CEST, MT & MRS
Neuro Thursday, 20 May 2021
Digital Poster

Oral Session - Imaging Metabolites: CEST, MT & MRS
Neuro
Thursday, 20 May 2021 12:00 - 14:00
  • Dynamic GlucoCEST MRI: results in primary brain tumors at 3 Tesla
    REGIS OTAVIANO FRANCA BEZERRA1, Hae Won Lee2, Gustavo Kaneblai3, Eduardo Figueiredo3, Mitsuharu Miyoshi4, Thomas Doring3, Claudia da Costa Leite5, Giovanni Guido Cerri2, and Frederico Perego Costa6
    1RADIOLOGY, HOSPITAL SÍRIO-LIBANÊS, SAO PAULO, Brazil, 2Radiology, Hospital Sírio-Libanês, Sao Paulo, Brazil, 3General Eletric, Sao Paulo, Brazil, 4General Eletric, Tokyo, Japan, 5RADIOLOGIA, HOSPITAL SÍRIO-LIBANÊS, SAO PAULO, Brazil, 6Oncology, Hospital Sírio-Libanês, Sao Paulo, Brazil
    The glucose signals measured by MTRasym and dynamic glucoCEST mean AUC values are significantly different in cancer and normal white matter in 3T for primary brain tumors
    Fig1.Neuroradiologist outlined 5 regions of interest (ROI)(A).Motion correction was done prior to B0 calculation. The signal % at 2 ppm was determined when B0 variation is lower than 1 ppm (B). The MTRasym at2 ppm represents the GlucoCEST signal (C). Dynamic DRY, INFUSION, CONTRAST acquired 20, 35 and 130 images respectively in 3 phases, each with 5 images, ranging from +3 to -1 ppm (D). Average of the 3 DRY phases generated a reference signal Sbase. INFUSION and CONTRAST acquired Sn images (E).
    Fig2.Mean MTRasym values from 15 patients with brain tumours were significantly higher in ROI 4 than in ROI 5 (ANOVA and TukeyHSD test) (A). The mean MTRasym values from ROI 1 to3 were significantly different from ROI 5 in the same 15 patients (ANOVA and Dunnetttest) (B).
  • Confounding of Macromolecular and Paramagnetic Tissue Content in Quantitative MTI Remedied by Explicit Estimation of Bound Pool Relaxation
    Alexey Samsonov1 and Aaron S. Field1
    1Radiology, University of Wisconsin-Madison, Madison, WI, United States
    Improved two-pool MT modeling with calibrated bound proton pool relaxation constraint may allow more specific assessment of macromolecular and paramagnetic tissue components. 
    Figure 2. Quantitative maps estimated using MT modeling with standard and proposed R1b in a healthy volunteer. (a) MPF and R1f. As predicted by simulations (Fig. 1b), standard R1f is dominated by macromolecular content and therefore resembles MPF. The proposed R1f is much more uniform, likely due to removing the effects of MPF. Note slightly elevated values in the basal ganglia, likely due to iron accumulation. (b) Error between the proposed and standard MPF. Note its high variability with macromolecular content and B1 field (c), also consistent with simulations (Fig. 1a).
    Figure 4. Quantitative mapping in an MS subject. (a) Proposed R1f is elevated in deep GM. The increase is consistent with the known distribution of iron in these regions, including areas with high (globus pallidus, #1), medium (putamen, #2), and low (thalamus, #3) iron content. Note elevated R1f in areas affected by diffuse lesional changes (#4) (as seen on T2-FLAIR and MPF). (b) The R1f is increased on the rim of the heavily demyelinated lesion (as revealed by MPF) (#1) and in WM areas affected by more diffuse disease (#2). (c) Example of a lesion without noticeable increase in R1f.
  • Improved volumetric inhomogeneous magnetization transfer (ihMT) using a CSF-suppressed FSE sequence (FLAIR-ihMT)
    Manuel Taso1, Fanny Munsch1, Olivier M Girard2, Guillaume Duhamel2, David C Alsop1, and Gopal Varma1
    1Division of MRI research, Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States, 2CRMBM, Aix-Marseille Univ, CNRS, Marseille, France
    We implemented a CSF-suppressed FSE sequence for myelin imaging using ihMT (FLAIR-ihMT). Simulations and experiments show the benefits of FSE imaging compared to gradient-echo, and the CSF suppression improves robustness of FSE imaging for ihMT myelin imaging in the CNS. 
    Figure 4 – 1.6mm high-resolution ihMT and ihMTR. A volume reconstructed with half-sampling is also shown
    Figure 3 – (a) ihMT-RAGE vs ihMT-FSE vs FLAIR-ihMT. Red arrows show area of improvement with FLAIR-ihMT compared to both ihMT-RAGE and FSE (b) comparison of ihMT image quality in infratentorial regions and (c) normalized ihMT signal comparison
  • Inhomogeneous magnetization transfer in the healthy adult brain: reproducibility and correlation with MTR and myelin water imaging
    Sarah Rosemary Morris1,2,3, Irene M. Vavasour1,4, Anastasia Smolina5,6, Erin MacMillan4,7, Guillaume Gilbert7, Michelle Lam2,4, Piotr Kozlowski1,2,4,8, Carl Michal2, Alan Manning2, Alex L. MacKay1,2,4, and Cornelia Laule1,2,4,8,9
    1Radiology, University of British Columbia, Vancouver, BC, Canada, 2Physics & Astronomy, University of British Columbia, Vancouver, BC, Canada, 3International Collaboration on Repair Discoveries, Vancouver, BC, Canada, 4UBC MRI Research Centre, Vancouver, BC, Canada, 5Physics & Astronomy, McMaster University, Hamilton, ON, Canada, 6Hospital for Sick Children, Toronto, ON, Canada, 7MR Clinical Science, Philips Healthcare Canada, Markham, ON, Canada, 8International Collaboration on Repair Discoveries (ICORD), Vancouver, BC, Canada, 9Pathology & Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
    Inhomogeneous magnetization transfer and myelin water imaging metrics (ihMTR and MWF) from white matter of healthy adults were compared. We found a moderately strong correlation between these two metrics and a good reproducibility for ihMTR.
    Figure 2: Example magnetization transfer ratio (MTR), inhomogeneous MTR (ihMTR) and myelin water fraction (MWF) maps from five of the participants. ihMTR and MWF show more variation in white matter than MTR.
    Figure 3: Pearson correlations between MTR and MWF and ihMTR and MWF. (ACR=Anterior corona radiata, AIC=Anterior limb of internal capsule, BOD=Body of corpus callosum, CEP=Cerebral peduncle, CIN=Cingulum, EXC=External capsule, GEN=Genu, PCR=Posterior corona radiata, PIC=Posterior limb of internal capsule, PTR=Posterior thalamic radiation, RIC=Retrolenticular part of internal capsule, SAS=Sagittal stratum, SCR=Superior corona radiata, SLF=Superior longitudinal fasciculus, SPL=Splenium)
  • GluCEST as an in vivo biomarker for monitoring abnormal glutamate dehydrogenase activity in Hyperinsulinism/Hyperammonemia syndrome at 7.0T
    Ravi Prakash Reddy Nanga1, Elizabeth A Rosenfeld2, Deepa Thakuri1, Mark Elliott1, Ravinder Reddy1, and Diva D De Leon2
    1Radiology, University of Pennsylvania, Philadelphia, PA, United States, 2Endocrinology and Diabetes, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
    Higher GluCEST contrast was observed on the lateral side of the hippocampus when compared to the contralateral side. The mean difference in GluCEST was 1.92% in nine subjects who have completed participation thus far.
    Figure 2: The top panel consists of an overlay of GluCEST map for the entire slice (left), followed by the overlay of only hippocampal ROIs (middle) and the corresponding T1map of the slice from MP2RAGE (right) from a male (top) and two female volunteers (center, bottom).
    Figure 1: GluCEST contrast values from the ROIs drawn on hippocampus of all the nine subjects as well as their rearranged values from hippocampus based on the side where GluCEST contrast was higher (termed as Lateral) and the other side (termed as Contralateral).
  • CEST Imaging of Nose-to-Brain Drug Delivery using Iohexol liposomes at 3T
    Lok Hin LAW1, Peng XIAO1, Jianpan Huang1, Xiongqi HAN1, and Kannie WY CHAN1,2,3
    1Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, Hong Kong, 2Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States, 3City University of Hong Kong Shenzhen Research Institute, Shenzhen, China
    In this study, we investigated the imaging of nanomedicine delivery via intranasal-administration using CEST-detectable mucus penetrating liposome. Ioh-Lipo generated CEST contrast of 33.4% at 4.3 ppm in phantom, which was also observed in-vivo in nostril, OB and FL after Injection at 3T.
    Fig.3 In vivo experiment of intranasal administration of 10%-PEG Iohexol Lipo (n=3). (A) Z-spectra, (B) CESTLDFit plots of CEST contrast at 4.3ppm, (C), T2 image of ICR mice brain, and (D), (E), (F), (G) CEST map Pre-Injection, and 0.5hr, 1hr, 1.5hr after intranasal injection at 4.3ppm.
    Fig. 1 CEST properties of 1%-PEG Ioh-Lipo and 10%-PEG Ioh-Lipo (n=3). (A) Z-spectra and (B) MTRasym of Ioh-Lipo under 0.9 μT and pH 7.0. (C) was the B1 power optimization on 10%-PEG Ioh-Lipo.
  • Peritumoral radiomics features from amide proton transfer-weighted MRI unveil the progressive pattern in early recurrent malignant gliomas
    Shanshan Jiang1, Pengfei Guo2, Hye Young Heo1, Peter van Zijl1,3, and Jinyuan Zhou1
    1Department of Radiology, Johns Hopkins University, Baltimore, MD, United States, 2Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, United States, 3F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, United States
     We explore radiomics features extracted from peritumoral areas on APTw images to unveil the progressive pattern in early recurrent malignant gliomas. Our results suggest that the use of APTw radiomic features can add important value to structural MRI to assess the treatment response.
    Fig. 1. Conventional MR images, APTw images and masks for two post-treatment GBM patients. Top case, a patient with tumor recurrence; bottom case, a patient with treatment effects.
    Fig. 2. CRT decision tree analysis on the basis of the data from FLAIR, APTw, and both.
  • Deuterium metabolic imaging of tumor burden and response to therapy in mutant IDH gliomas in vivo
    Celine Taglang1, Georgios Batsios1, Mers Tran1, Anne Marie Gillespie1, Hema Artee Luchman2, Russell O Pieper3, Sabrina M Ronen1, and Pavithra Viswanath1
    1Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, United States, 2Cell Biology and Anatomy, University of Calgary and Hotchkiss Brain Institute, Calgary, AB, Canada, 3Neurological Surgery, University of California San Francisco, San Francisco, CA, United States
    Here, we show that [6,6’-2H]-glucose non-invasively monitors tumor burden and response to therapy in preclinical low-grade glioma models at early timepoints prior to alterations in tumor volume, pointing to its potential to assess pseudoprogression, which is a challenge in glioma imaging.
    Figure 3. [6,6’-2H]-glucose flux to lactate is localized to the tumor region in low-grade gliomas in vivo. Representative metabolic heatmaps from 2D CSI studies in mice bearing orthotopic BT257 tumor xenografts following injection of a bolus of [6,6’-2H]-glucose. Left panel shows a representative axial T2-weighted MR image while the right panel shows a metabolic heatmap of the SNR of lactate produced from [6,6’-2H]-glucose.
  • CRT-FID-MRSI at 7T for the high-resolution metabolic imaging of epilepsy: Preliminary results
    Gilbert Hangel1, Philipp Lazen2, Matthias Tomschik1, Jonathan Wais1, Eva Hečková2, Lukas Hingerl2, Stephan Gruber2, Bernhard Strasser2, Gregor Kasprian3, Daniela Prayer3, Julia Furtner3, Christoph Baumgartner4, Johannes Koren4, Robert Diehm5, Martha Feucht5, Christian Dorfer1, Ekaterina Pataraia6, Wolfgang Bogner2, Siegfried Trattnig2,7, and Karl Rössler1
    1Department of Neurosurgery, Medical University of Vienna, Vienna, Austria, 2High Field MR Centre, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria, 3Division of Neuroradiology and Musculoskeletal Radiology, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria, 4Department of Neurology, Clinic Hietzing, Vienna, Austria, 5Department of Paediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria, 6Department of Neurology, Medical University of Vienna, Vienna, Austria, 7Christian Doppler Laboratory for Clinical Molecular MR Imaging, Vienna, Austria
    We show the feasibility of CRT-FID-MRSI at 7T for high resolution metabolic imaging in epilepsy. We acquired a more extensive metabolic profile at higher resolution than previous MRSI, with tCho and tCr/NAA/mIns/Glu as most promising markers.
    Figure 3: Overview of the possible 3D lesion localisation for multiple metabolites in patient #2, an FCD 1b patient. The hotspot location corresponds well to later focal resection.
    Figure 5: Metabolic ratio maps overlayed to post-resection clinical imaging shows good correspondence of 7T metabolic imaging to surgical FCD resection.
  • In vivo GABA increase as a biomarker of the epileptogenic zone: an unbiased metabolomics approach
    Florence Fauvelle1,2, Vasile Stupar1,2, Jia Guo3, Wafae Labriji1, Chen Liu3, Alicia Plaindoux1, Emmanuel Luc Barbier1,2, Sophie Hamelin1, and Antoine Depaulis1
    1Grenoble Institut Neurosciences, University Grenoble Alpes, La Tronche, France, 2IRMaGE, University Grenoble Alpes, La Tronche, France, 3Departement of Psychiatry, Columbia University, New York, NY, United States
    Using ex vivo NMR spectroscopy-based (MRS) untargeted metabolomics and in vivo edition-MRS method, we demonstrated that GABA was a robust in vivo biomarker of epileptogenic zone in mesio-temporal lobe epilepsy.
    Figure 3: A/ In vivo GABA edition using MEGA-PRESS pulse sequence. Examples of GABA and GLX peaks in the subtracted spectrum, fitted using the JET algorithm, in Sham (top) and KA-MTLE (bottom) mice (regular KA dose). B/ Regression curve between in vivo GABA+/GLX and ex vivo GABA/GLX
    Figure 2: A/ Score plot of the principal component analysis (PCA) built with HRMAS MRS data of KA-MTLE mice and the 5 brain regions sampled (ipsilateral and contralateral hippocampus (IH and CH), posterior (P) and anterior (A), and adjacent cortex) . B/ Variable importance in the projection (VIP) ranking metabolites from most discriminative (VIP>1) to less discriminative ones in KA-MTLE mice C/ Percentage of variation of 8 metabolites (among the 19 metabolites quantified) in the 5 brain regions of KA-MTLE mice vs their sham homologous.
Back to Top
Digital Poster Session - Imaging Metabolites: CEST & MT
Neuro
Thursday, 20 May 2021 13:00 - 14:00
  • Whole-brain B1-corrected quantitative MT imaging in less than 5 minutes
    Roya Afshari1,2, Francesco Santini1,2, Rahel Heule3,4, Craig Meyer5, Josef Pfeuffer6, and Oliver Bieri1,2
    1Department of Radiology, Division of Radiological Physics, University Hospital Basel, Basel, Switzerland, 2Department of Biomedical Engineering, University of Basel, Basel, Switzerland, 3Department of High Field Magnetic Resonance, Max Planck Institute for Biological Cybernetics, Tübingen, Germany, 4Department of Biomedical Magnetic Resonance, University of Tübingen, Tübingen, Germany, 5Department of Biomedical Engineering, University of Virginia, Virginia, VA, United States, 6Department of Application Development, Siemens Healthcare, Erlangen, Germany
    Using a spiral prototype sequence, accurate whole brain B1-corrected quantitative magnetization transfer (qMT) imaging is feasible within less than 5 minutes in the clinical setting at 3T.
    Figure 2: Illustrative axial views at three positions for all the retrieved parameter maps. From left to right: the bound pool fraction (F), the exchange rate (kf), the transverse relaxation time of the bound pool protons (T2r), the observable longitudinal relaxation time of the free pool protons (T1,obs), and the B1-map. Yellow and red ROIs shown on the middle row of the F slices were used for the data presented in Table 1.
    Table 1: Mean and standard deviation of the bound pool fraction (F), the exchange rate (kf), the transverse relaxation time of the bound pool protons (T2r), the observable longitudinal relaxation time of the free pool protons (T1,obs) calculated over ROIs selected in white matter (red rectangle) and gray matter (yellow rectangle) shown in the middle row of F slices in Figure 2.
  • Insignificant contribution of blood to NOE(-1.6)
    Jing Cui1, Yu Zhao1, Feng Wang1, Junzhong Xu1, Daniel Gochberg1, John Gore1, and Zhongliang Zu1
    1Vanderbilt University Medical Center, Nashville, TN, United States
    We used signal acquisition with a diffusion-weighting and injection of MION to evaluate the contribution of blood to NOE(-1.6) in rat brains. Results suggest that NOE(-1.6) is not mainly from blood, and that MION particles alter the NOE(-1.6) but have much weaker effects on other CEST and NOE.
    Fig. 1. Averaged Z-spectra and AREX spectra for each pool (a, b), statistics of fitted AREX values for amide (c), NOE(-1.6) (d), and NOE(-3.5) (e) as well as S0 (f) from the whole brain with a diffusion-weighting of b = 0s/mm2 (blue) and 400s/mm2 (red), respectively. Each AREX spectrum contains a CEST or NOE peak from a corresponding pool and a residual water peak. The great residual water peak is due to the use of the inverse analysis for calculating AREX values. * P < 0.05
    Fig. 3. Averaged Z-spectra and AREX spectra for each pool (a, b), statistics of fitted AREX values for amide (c), NOE(-1.6) (d), and NOE(-3.5) (e) as well as S0 (f) from the whole brain before (blue) and after (red) the injection of 5 mg/kg MION, respectively. * P < 0.05
  • The Effect of Cariporide on Tumour Intracellular pH: A Study in Rat C6 Glioma using AACID-CEST-MRI
    Maryam Mozaffari1,2, Nivin Nystrom1,2, Alex Li1, Miranda Bellyou1, Timothy Scholl1,2, and Robert Bartha1,2
    1Robarts Research Institute, London, ON, Canada, 2Department of Medical Biophysics, Western University, London, ON, Canada
    The intracellular pH of rat C6 glioma was measured at two time-points and found to be relatively basic compared to contralateral tissue. Cariporide did not selectively acidify this model as previously observed in mouse U87MG tumours.
    Fig. 2: a) Representative anatomical image (T2-weighted) and superimposed colour-coded AACID maps for b) baseline and c) 60 minutes after injection of cariporide at day 14 post-implantation of the tumour. The tumour region is highlighted by the purple line. The average AACID value in tumour regions pre- and post-injection of cariporide was 1.22±0.02 and 1.27±0.020, respectively.
    Fig. 1: The average AACID values for a) control groups (N=15) in right and left frontal lobes. b) experimental groups at 7-9 days (N=22) and 14-16 days (N=20) post tumour implantation. Error bars represent the standard error of the mean. The asterisks indicated p<0.05 in the paired t-test.
  • Numerical Fit of Extrapolated Semisolid Magnetization Transfer Reference Signal (NEMR) for Improved pH-Weighted Imaging of Ischemic Stroke
    Xingwang Yong1, Shanshan Lu2, Yi-Cheng Hsu3, Yi Sun3, Dan Wu1, and Yi Zhang1
    1Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, Zhejiang, China, 2The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China, 3MR Collaboration, Siemens Healthcare Ltd., Shanghai, China
    A novel method was proposed for removing contamination in the CEST signal by the numerical fitting of the extrapolated semisolid magnetization transfer reference signal (NEMR). NEMR yielded substantially better contrast for depicting ischemic stroke lesions than the MTRasym method.
    Figure 2. DWI, APTw, APT#, and NOE# images of a representative case using saturation B1 power of 1uT and 1.5uT.
    Figure 5. DWI, APTw, APT#, and NOE# images of a representative case at a B1 power of 1uT.
  • Promising nerve imaging biomarkers for applications in inherited neuropathies
    Alison R Roth1, Jun Li2, and Richard Dortch1
    1Neuroimaging Research, Barrow Neurological Institute, Phoenix, AZ, United States, 2Neurology, Wayne State University, Detroit, MI, United States
    Magnetization transfer ratio (MTR) from the sciatic nerve were found to be the most promising potential imaging biomarkers in patients with inherited neuropathies. Cross-sectional area, circularity, eccentricity, and nerve fascicle density also showed potential.
    Figure 2: Box and whisker plots with data points of MTR (magnetization transfer ratio), CSA (cross-sectional area), circularity, eccentricity, and nerve fascicle density by subject type. CMT1A is Charcot-Marie-Tooth Type 1A, CMT2A is CMT type 2A, and HNPP is hereditary neuropathy with liability to pressure palsy.
    Figure 1 Left: Representative magnetization transfer (MT)-weighted MRI images of study subjects with segmented sciatic nerves in red in magnified inlay. Right: Magnetization Transfer Ratio (MTR) for subjects. Subjects in row A are healthy controls, B have Charcot-Marie-Tooth type 1A, C have CMT type 2A, and D have hereditary neuropathy with liability to pressure palsy (HNPP). Patient imaging values are shown in the table.
  • Using Glutamate-Weighted MR Imaging (GluCEST) to Detect Effects of Transcranial Magnetic Stimulation
    Abigail T.J. Cember1, Benjamin Deck2, Jared Zimmerman3, Brian Erickson2, Apoorva Kelkar2, Olufunsho Faseyitan3, Mark Elliott1, Ravinder Reddy1, and John D. Medaglia2
    1Center for Magnetic Resonance and Optical Imaging, Department of Radiology, University of Pennsylvania, Philadelphia, PA, United States, 2Department of Psychology, Drexel University, Philadelphia, PA, United States, 3Laboratory for Cognition and Neural Stimulation, Department of Neurology, University of Pennsylvania, Philadelphia, PA, United States
    We find that healthy volunteers undergoing continuous theta burst stimulation (cTBS), a form of transcranial magnetic stimulation (TMS) to the left motor cortex (M1) exhibit a statistically significant, non-localized decrease in the gluCEST signal measured at ultra-high field (7T). 

    Figure 1. GluCEST maps: data from all subjects projected onto the anatomy of a single subject for visual representation. A) average gluCEST by segment, baseline (pre-stimulation) -15 subjects. B) post "sham" (placebo) stimulation - 5 subjects. C) post cTBS (real) stimulation -- 10 subjects. The colorscale is identical in all maps, with settings as shown in the screenshot from ITK-SNAP. Green arrow indicates the left precentral gyrus, the intended target of cTBS in stimulated subjects. Please see main text for full listing of anatomical segments treated distinctly in this analysis.

    Figure 2. Barplot of gluCEST changes by segment: 99% confidence intervals (CI) of mean change, as report by unpaired T-test. A cluster of four bars is shown for each segment as listed in the text, beginning with the Precentral Gyrus for each side. A T-test was performed to estimate the 99% confidence interval for the change in each segment for both the sham and stimulated subjects. The 'true' change likely lies between the two confidence intervals, generally giving a value near zero for sham subjects, but a small negative value for stimulated subjects.
  • A digital human head phantom for validation of retrospective motion correction in glucoCEST MRI
    Patrick M. Lehmann1, Mads Andersen2, Anina Seidemo1, Xiang Xu3,4, Xu Li4,5, Nirbhay Yadav4,5, Ronnie Wirestam1, Frederik Testud6, Patrick A. Liebig7, Pia C. Sundgren8,9, Peter C. M. van Zijl4,5, and Linda Knutsson1,4
    1Department of Medical Radiation Physics, Lund University, Lund, Sweden, 2Philips Healthcare, Copenhagen, Denmark, 3BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States, 4Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States, 5F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, United States, 6Siemens Healthcare AB, Malmö, Sweden, 7Siemens Healthcare GmbH, Erlangen, Germany, 8Department of Radiology, Lund University, Lund, Sweden, 9Lund University Bioimaging Centre, Lund University, Lund, Sweden
    A digital human head phantom can be used to reproduce motion artefacts reported in in vivo glucoCEST images and to analyse and validate motion correction approaches with respect to the truth of the residual contrast.
    Figure 2: Illustration of a glucoCEST scan of a patient with a brain tumour under the influence of D-glucose infusion, rigid-head motion and dynamic lateral ventricle dilatation and contraction.
    Figure 4: AUC (area under the curve) maps depicting different intervals: Pre-infusion (115 s), during infusion (245 s), and two post-infusion time intervals (45 s before, 160 s after peak) showing signal rise and decay. Five cases showing ground truth with D-glucose infusion and without motion (E), without infusion and with motion (A, C), and with infusion and motion (B, D), before (A, B) and after (C, D) retrospective motion correction.
  • Volumetric glutamate-weighted MR imaging (gluCEST) enables in vivo detection of metabolic differences between human hippocampal subfields
    Abigail T.J. Cember1, Ravi Prakash Reddy Nanga1, Hari Hariharan1, Neil E. Wilson2, Puneet Bagga3, and Ravinder Reddy1
    1Center for Magnetic Resonance and Optical Imaging, Department of Radiology, University of Pennsylvania, Philadelphia, PA, United States, 2Siemens Medical Solutions, USA, Malvern, PA, United States, 3Department of Diagnostic Imaging, St. Jude Children's Research Hospital, Memphis, TN, United States
    We performed volumetric (3D) gluCEST to image a slab containing the medial temporal lobe (MTL) in healthy subjects. Upon regional analysis, we find amongst other trends that the dentate gyrus has greater gluCEST contrast than neighboring regions.
    Figure 4. Example gluCEST map through medial temporal lobe slice, shown in transparent overlay on the T2-weighted structural image.
    Figure 3. Comparison of gluCEST values in left and right hippocampal subfields. This boxplot is analogous to those in Figure 1, although rather than combining the corresponding measurements on the left and right sides of the brain, we inspect each separately. It can be seen from this plot that the gluCEST distribution in the dentate gyrus is higher than in the other subfields to a corresponding degree on both sides of the brain.
  • Automated CEST Measurements for the Lateralization of Epileptic Foci in Temporal Lobe Epilepsy at 3 T
    Qingqing Wen1, Kang Wang2, Wenqi Wang1, Yi Sun3, Dan Wu1,2, and Yi Zhang1,2
    1Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, China, 2Department of Neurology, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China, 3MR Collaboration, Siemens Healthcare Ltd., Shanghai, China
      Automated CEST measurements in the hippocampus and amygdala at 3T can improve the diagnostic accuracy for the seizure lateralization of TLE patients, compared with conventional quantitative T1 and T2 mapping.
    Figure 4. Coronal slices of the reconstructed T1w images (A, E), mean MTRasym maps within 2-4 ppm (B, F), T1 maps (C, G), and T2 maps (D, H) of a left TLE patient (first row) and a right TLE patient (second row). The MTRasym values were higher in the epileptogenic hippocampus and amygdala (HA, illustrated by red boxes) than those in the contralateral HA. However, T1 and T2 values in the HA ipsilateral to the seizure were similar to or even lower than those in the contralateral HA (D, G, and H).
    Figure 3. ROC curves for the lateralization of epileptic foci using metric1 (A) and metric2 (B). The two metrics used MTRasym (blue line), T1 (red line), and T2 (yellow line) values as input indices. For both of these two metrics, AUC values of the MTRasym index were significantly higher than those of T1 and T2.
  • ihMT Analysis of Myelin in the Shiverer Mouse Brain
    Choong Heon Lee1, Piotr Walczak2, and Jiangyang Zhang1
    1Radiology, NYU Medical Center, New York, NY, United States, 2University of Maryland School of Medicine, Baltimore, MD, United States
    ihMT can detect residual non-compact myelin in the context of cellular infiltrates in the dysmyelinating shiverer mouse model, suggesting higher sensitivity to myelin than conventional MT.
    Fig. 5: Representative images of control and shiverer mouse brains. T2-weighted image (leftmost) is compared with ihMTR image at a dual offset frequency of 12 kHz as well as MTR image at offset frequencies of 4, 8, 16 kHz. Corpus callosum (orange arrow), cerebral peduncle (yellow), and trigeminal nerve (green).
    Fig. 3: Optimization of ihMTR signals in shiverer mouse brains (n=3). A series of dual offset frequencies ranging from 4k to 24k in horizontal axis; time delays from 0.01 ms to 4 ms in vertical axis; and saturation power in diagonal axis.
  • The value of Amide Proton Transfer weighted and Dynamic Contrast-Enhanced imaging in peritumoral edema assisted grading of gliomas
    Xinying Ren1, Yujing Li1, Rui Wang1, Tao Wen1, Diaohan Xiong1, Pengfei Wang1, Guangyao Liu1, Jing Zhang1, and Kai Ai2
    1Department of Magnetic Resonance, Lanzhou University Second Hospital, Lanzhou, China, 2Philips Healthcare, Xi'an, China
    The APT value and the DCE-MRI parameters of Ktrans  of peritumor edema may serve as a potential biomarker assisted grading of gliomas. The former may be more conductive to differentiate grade Ⅱ and grade Ⅲ gliomas.
    *Note: Using the one-way ANOVA and spearman’s correlation coefficient for ranked data
    Figure 1. ROC curve of APT value and Ktrans value in peritumoral edema between grade Ⅱ and Ⅲ (AUC: 0.971 for APT, 0.714 for Ktrans; 95%CI: 0.89-1.00 for APT, 0.41-1.00 for Ktrans).
  • Discrimination of IDH1 Genotype and 1p/19q Status in Glioma: A comparison study between Arterial Spin Labeling and Amide Proton Transfer imaging
    DiaoHan Xiong1, Rui Wang1, Tao Wen1, Yujing Li1, Xinying Ren1, Pengfei Wang1, Guangyao Liu1, Jing Zhang1, and Kai Ai2
    1Department of Magnetic Resonance, Lanzhou University Second Hospital, Lanzhou, China, 2Philips Healthcare, Xi'an, China
    Both APTw and 3D-pCASL have the predictive value of IDH1 Genotype and 1p/19q Status before surgery, the 3D-pCASL is btter. It may be helpful for method choosing and clinical management .
    Fig 4: Two representative cases for gliomas with aligned Contrast enhancement(A,D), APTw (B,E) and 3D-pCASL.A 48-year-old female with 1p/19q codeletion type (A,B,C),shows low APTw value and low perfusion. A 58-year-old male, with 1p/19q undeletion (E,F,G),shows high APTw value and high perfusion.
    Fig 2: ROC curve for APTw and 3D-pCASL in distinguishing IDH mutation type and IDH wild type. ROC curve: receiver operating characteristic curve
  • Potential feasibility of new parameters on CEST imaging by multi pool model in relation to 11C-MET uptake on PET/CT and IDH1 mutation in gliomas
    Yasukage Takami1, Naruhide Kimura1, Katsuya Mitamura1, Takashi Norikane1, Keisuke Miyake2, Tatsuya Yamasaki3, Kazuo Ogawa3, Mitsuharu Miyoshi4, and Yoshihiro Nishiyama1
    1Department of Radiology, Faculty of Medicine, Kagawa University, Miki-cho, Japan, 2Department of Neurological Surgery, Faculty of Medicine, Kagawa University, Miki-cho, Japan, 3Department of Clinical Radiology, Kagawa University Hospital, Miki-cho, Japan, 4Global MR Applications & Workflow, GE Healthcare Japan, Hino-shi, Japan
    Several correlation coefficients between MTRasymmax and T/N ratio, MTRasymmean and T/N ratio, APT_T1max and T/N ratio, and APT_T1mean and T/N ratio were relatively high (r = 0.38, 0.46, 0.49, and 0.53, respectively), but not statistically significant.
    Fig.1 Correlation between parameters on CEST imaging by multi pool model and MET T/N ratio. The Pearson's test showed correlation between MTRasymmean and MET T/N ratio (a) (r = 0.46), APT_T1mean and MET T/N ratio (b) (r = 0.53)
    Fig.2 Comparison of average MTRasymmean (a), APT_T1mean (b) and MET T/N ratio (c) for IDH1-mutant glioma patients, IDH1 wildtype patients (* p<0.05).
  • Amide proton transfer-weighted MR imaging in the rat brain of demyelination and remyelination
    Do-Wan Lee1, Hwon Heo2, Chul‐Woong Woo3, Jae-Im Kwon3, Joongkee Min3, Monica Young Choi2, Yeon Ji Chae2, Dong‐Cheol Woo2,3, Kyung Won Kim1, Jeong Kon Kim1, Hyo Jeong Chin4, and Dong‐Hoon Lee4
    1Department of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea, Republic of, 2Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea, Republic of, 3Convergence Medicine Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Korea, Republic of, 4Department of Radiological Science, College of Health Sciences, Yonsei University, Wonju, Korea, Republic of
    Amide proton transfer-weighted (APTw) 7T MRI allows the monitoring of changes in amide proton concentration levels in the brain regions of demyelination and remyelination.
    Figure 3. Reconstructed and overlaid amide proton transfer-weighted (APTw) image of the whole-brain and the corpus callosum in an unsaturated image of a typical rat in each group (control group, normal control group; DEM, demyelinated group; and REM, remyelinated group). The black lines in the images of the whole-brain area for all groups indicate the contour of the corpus callosum. The color bar represents the range of calculated APTw signal (%).
    Figure 2. Averaged Z-spectra (a), magnified Z-spectra from 0 to 6 ppm (b), magnified Z-spectra from -6 to 0 ppm (c) , magnetization transfer ratio asymmetry (MTRasym) curves (d), and amide proton transfer-weighted (APTw) signals calculated in the corpus callosum at each group (e) (Msat, signal intensity with RF saturation; M0, signal intensity without RF saturation; controls, black; DEM, demyelinated group, red; and REM, remyelinated group, green). **p = .009 and ***p < .001.
  • CEST and AREX data processing based on deep neural network: application to image Alzheimer’s disease at 3T
    Jianpan Huang1, Joseph H. C. Lai1, Kai-Hei Tse2, Gerald W.Y. Cheng2, Xiongqi Han1, Yang Liu1, Zilin Chen1, Lin Chen3,4, Jiadi Xu3,4, and Kannie W. Y. Chan1,4,5
    1Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China, 2Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China, 3F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Research Institute, Baltimore, MD, United States, 4Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States, 5City University of Hong Kong Shenzhen Research Institute, Shenzhen, China
    Deep neural network based CEST/AREX were exploited to analyze CEST data of mouse brains with Alzheimer’s disease (AD). Significant lower CEST/AREX signals related to amyloid β-peptide  plaques were detected in AD mouse brains compared to age-matched WT mouse brains.
    Figure 1. The schematics of CESTNet (A) and AREXNet (B).
    Figure 3. Representative CEST and AREX maps of WT and AD brains, generated by trained CESTNet and AREXNet. CEST maps (AAPT, ArNOE and AMT) of central (A) and anterior slice (B). AREX maps (RAPT, RrNOE and RMT) of central slice (C) and anterior slice (D).
  • CEST imaging with neural network fitting of the human brain at 3T
    Zhichao Wang1, Yu Zhao2, Xu Yan3, Zhongshuai Zhang3, Caixia Fu3, Hui Tang4, and Jianqi Li1
    1Shanghai Key Laboratory of Magnetic Resonance, School of Physics and Electronic Science, East China Normal University, Shanghai, China, 2Institute of Imaging Science, Vanderbilt University, Nashville, TN, United States, 3MR Collaboration NE Asia, Siemens Healthcare, Shanghai, China, 4Department of Radiology, Renji Hospital affiliated to Shanghai Jiao Tong University Medical College, Shanghai, China
     Separating different targets is highly valuable for clinical application of CEST. In this study, the background Z-spectra including only the magnetization transfer and direct saturation effects was fitted by using neural network, then CEST and NOE maps were obtained simultaneously. 
    FIGURE 1 Schematic of data processing pipeline. Simulated background Z-spectrum (A) are generated. In each simulated background Z-spectrum (C), the data marked as red solid dots are inputted for training and the data in blue line are target for training. (D)The feedforward neural network. (B) The data marked as red solid dots from the acquired Z-spectrum are inputted for prediction. (E) The background Z-spectrum (marked as dashed blue curve) is obtained from the network. (F)The APT map and NOE map are obtained by subtracting background Z-spectrum from the acquired Z-spectrum.
    FIGURE 3 The results from a patient with cerebral infarction. The 1st row includes the conventional T1-weighted image (A), T2-weighted FLAIR image (B), APT map (C) and NOE map (D). The 2nd row includes the fitting Z- spectrum of infarction tissue (E) and normal tissue (F), boxplots of APT contrast (G) and NOE contrast (H) between infarction and normal tissues. In (E) and (F), the red curves are B0-corrected Z- spectra, and the blue lines are background Z- spectra from neural network fitting. The red arrow in the T2W FLAIR image indicates the area of the lesion.
  • Differentiation of Radiation Necrosis from Tumor Progression in Brain Metastasis Treated with Stereotactic Radiosurgery using CEST at 3T
    Rachel W Chan1, Hatef Mehrabian1, Hany Soliman2, Hanbo Chen2, Aimee Theriault2, Sten Myrehaug2, Chia-Lin Tseng2, Jay Detsky2, Wilfred W Lam1, Angus Z Lau1,3, Gregory J Czarnota1,2,3, Arjun Sahgal2, and Greg J Stanisz1,3,4
    1Physical Sciences, Sunnybrook Research Institute, Toronto, ON, Canada, 2Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, ON, Canada, 3Medical Biophysics, Sunnybrook Research Institute, Toronto, ON, Canada, 4Department of Neurosurgery and Pediatric Neurosurgery, Medical University, Lublin, Poland
    CEST at 3T can be used to distinguish between radiation necrosis and tumor progression after stereotactic radiosurgery. The amide MTR parameter acquired at 0.52μT was selected from multivariable modelling with an AUC of 0.91.
    Figure 5 – Tumor and Radiation Necrosis: (A) The median values with violin plots are shown for the tumor (red) and radiation necrosis (green) outcomes. Asterisks represent significant differences (**p<0.01, *p<0.05) between the two outcome groups after adjusting for multiple testing. (B) The ROC curve is shown of the significant parameter after multivariable modelling for predicting a tumor outcome.
    Figure 3 – Example of Radiation Necrosis: The post-gadolinium T1-weighted images are shown along with the four CEST maps – MTR amide and rNOE, acquired with B1=0.52μT and B1=2.0μT. The values on the bottom right represent the ROI medians and standard deviations.
  • Differentiating PD and MSA-P with Neuromelanin and Iron Simultaneously Using a Single 3D Magnetization Transfer Sequence
    Yu Liu1, Junchen Li2, Ying Wang3, Naying He1, Zhijia Jin1, Pei Huang4, Shengdi Chen4, Fuhua Yan1, and Ewart Mark Haacke3
    1Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China, 2Department of Radiology, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, Changshu, China, 3Wayne State University, Detroit, MI, United States, 4Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
    In this study, we observed that MSA with a parkinsonian variant had significantly smaller neuromelanin (NM) volume compared with Parkinson’s disease (PD) patients (p=0.0017) and healthy controls (HCs) (p=0.0013).

    Data are presented as median ± interquartile range unless otherwise noted.

    Fig. 1. MTC magnitude images for an MSA-P patient, PD patient and HC are shown. From the MTC magnitude images of the MSA-P and PD patients, we can easily observe the NM depigmentation with lower contrast-to-noise than HC. The patients with MSA-P apparently suffer the worst NM degeneration.
  • Comparison of Capability for Molecular-Based Assessment between 3D Gradient Echo-Based and 2D Spin Echo-Based CEST Imaging for Brain Tumors
    Kazuhiro Murayama1, Yoshiharu Ohno2, Masao Yui3, Kaori Yamamoto3, Masato Ikedo3, Satomu Hanamatsu2, Akiyoshi Iwase4, Takashi Fukuba4, and Hiroshi Toyama2
    11) Joint Research Laboratory of Advanced Medical Imaging, Fujita Health University School of Medicine, Toyoake, Japan, 2Radiology, Fujita Health University School of Medicine, Toyoake, Japan, 3Canon Medical Systems Corporation, Otawara, Japan, 4Radiology, Fujita Health University Hospital, Toyoake, Japan
    3D chemical exchange saturation transfer (CEST) imaging has equal to or higher potentials for molecular-based assessment and can be considered at least as valuable as 2D CEST imaging in patients with various brain tumors.

    Figure 1. 50-year old male with glioblastoma (Grade Ⅳ).

    Glioblastoma (arrow) is shown in the left temporal cerebral suubcortical and white matter on Fluid-attenuated inversion recovery (FLAIR), Contrast-enhanced T1-weighted imaging (CE-T1WI) and CEST imaging. 3D CEST imaging can cover the whole lesion as compared with 2D CEST imaging. MTRasym at 3.5ppm in this case were 2.12% (2D CEST imaging) and 1.69% (3D CEST imaging). 2D and 3D CEST imaging were diagnosed as glioblastoma (grade Ⅳ).

    Figure 2. Results of correlation and Bland-Altman plot analysis between 2D and 3D CEST imaging measurement of MTRasym (at 3.5 ppm) for all lesions.

    3D CEST imaging had significant and excellent correlation with 2D CEST imaging (r=0.80, p<0.0001). The limits of agreement between 2D and 3D CEST imaging was -0.012±0.73 (mean±1.96 × standard deviation) %.

  • Prediction of the response to induction chemotherapy using Amide Proton Transfer MRI in nasopharyngeal carcinoma
    Guixiao Xu1, Hui Li1, Yueming Yuan2, Liangru Ke1, Yun He1, Yanlin Zhu1, Liyun Zhen2, Yingyi Huang1, Chuanmiao Xie1, and Yongming Dai2
    1Department of Radiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in Southern China, Guangzhou, China, 2Central Research Institute, United Imaging Healthcare, Shanghai, China
    We found that the pre-treatment mean APT value was correlated to percentage change of primary tumor size negatively in NPC. This indicated the NPC pre-treatment APT value might be a useful features or biomarker of the response in IC.
    Figure 2. Pre-treatment axial APTw image of a primary nasopharyngeal carcinoma in a 44-year-old woman (APTmean = 1.34%) and a 26-year-old woman (APTmean = 2.64%).
    Figure 1. The correlation between mean APT value (APTmean) and percentage change (%Δ area) of primary tumor.
Back to Top
Digital Poster Session - Imaging Metabolites in the Brain
Neuro
Thursday, 20 May 2021 13:00 - 14:00
  • Fast High-Resolution 1H-MRSI of the Human Brain at 7T
    Rong Guo1,2, Yibo Zhao1,2, Yudu Li1,2, Pallab Bhattacharyya3, Mark Lowe3, Hannes M. Wiesner 4, Yao Li5, Xiao-Hong Zhu4, Wei Chen4, and Zhi-Pei Liang1,2
    1Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, United States, 2Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, United States, 3Imaging Institute, Cleveland Clinic, Cleveland, OH, United States, 4Center for Magnetic Resonance Research, University of Minnesota, Minneapolis, MN, United States, 5School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
    A rapid, high-resolution, non-water-suppressed MRSI technique was developed at 7T. Using the SPICE framework, the new technique can simultaneously obtain metabolite maps at 3.0×3.0×3.0 mm3 resolution and water signals at 1.0×1.0×3.0 mm3 resolution in an 8 min scan with high fidelity.
    Figure 4. Representative metabolite maps (NAA, Cr, Cho) and spatially localized spectra obtained from a healthy subject using SPICE. High-quality spatiospectral distributions of various brain metabolites were obtained.
    Figure 5. Simultaneously obtained water image, QSM map, T2* map at 1.0×1.0×3.0 mm3 nominal resolution and metabolite maps of NAA, Cr and Cho from two representative traverse images at 3.0×3.0×3.0 mm3 nominal resolution from a healthy subject in a single 8-min scan using SPICE.
  • GABA Inhibition Enhances in Epilepsy Associated with Focal Cortical Dysplasia
    Tao Gong1, Yufan Chen1, Liangjie Lin2, Youting Lin3, and Guangbin Wang1
    1Shandong Medical Imaging Research Institute, Shandong University, Jinan, China, 2Philips Healthcare, Beijing, China, 3Departments of Neurology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
    The results of this study indicated that GABAergic inhibition enhanced in FCD foci of epilepsy patients, while no significant alteration of GSH and Glx levels was found, suggesting GABA may play a central role in the pathophysiology of FCD-associated epilepsy.
     
    Figure 1. T2-FLAIR image (a) of an FCD (white arrow) -associated epilepsy patient, the volumes of interest of HERMES in the patient (b and c) and in a matched healthy control (d). The mean (± standard deviation) GABA (e), Glx (e) and GSH (f) -edited spectra from the HERMES sequence in FCD foci, contralateral regions and healthy controls.
    Figure 2. Comparation of GABA, GSH and Glx levels between the FCD foci, contralateral regions and healthy controls using ANOVA, and the results indicated that GABA levels was significantly increased in FCD foci compared with contralateral regions (p=0.007) and with healthy controls (p=0.003).
  • Multimodal 1H-MRSI and 18F-FDG-PET in Temporal Lobe Epilepsy
    Lihong Tang1, Hui Huang1, Miao Zhang2, Yibo Zhao3,4, Rong Guo3,4, Yudu Li3,4, Zhi-Pei Liang3,4, Wei Liu5, Yao Li1, Biao Li2, and Jie Luo1
    1School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China, 2Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China, 3Department of Electrical and Computer Engineering, University of Illinois at Urbana Champaign, Urbana, IL, United States, 4Beckman Institute for Advanced Sciences and Technology, University of Illinois at Urbana Champaign, Urbana, IL, United States, 5Department of Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
    Simultaneous high-resolution 1H-MRSI (2.0 x 3.0 x 3.0 mm3), MWF map, QSM map, and 18F-FDG-PET offer complementary information in subcortical, cortical and white matter regions for drug-resistant TLE patients, demonstrating potential for lateralization of epileptogenic zone.
    Figure 1. The coronal view high-resolution multimodal imaging of patient #3, including T1w-MPRAGE, PET-SUVR, NAA, Cr, Cho, Ins, NAA/Cr, NAA/(Cho+Cr), MWF, and QSM maps. Red arrows point to the ipsilateral temporal lobe.
    Figure 4. Relationships between A) NAA/(Cho+Cr) and PET-SUVR in hippocampus; B) NAA/(Cho+Cr) and PET-SUVR in thalamus; C) NAA/(Cho+Cr) and MWF in cingulum hippocampus. P and r values are results of Pearson’s correlation.
  • Dopamine Directional Circuits Detected by Metabolic Effective Connectivity and Granger Causality using Integrated PET/MR
    Lei Wang1, Longxiao Wei1, and Menghui Yuan1
    1Nuclear Medicine, Tangdu Hospital of Air Force Medical University, Xi'an, China
    Metabolic effective connection method based on integrated PET/MR technology gives more consistent directional pathways than granger causality. Metabolic effective connection is more suitable for region-wise study.
    Fig3. The group-wise directional pathways identified by Metabolic effective connection(MEC) method. Arrows represent the direction of the pathways. The line thickness represents the relative strength of the MEC. There were more bidirectional interactions between NAc and caudate, and the unidirectional pathway from OFC to caudate revealed the regulation in frontostriatal dopamine pathway. (One-sample t-test, p<0.05, fdr correted)
    Fig2. The group-wise directional network identified by Granger causality. Arrows represent the direction of the pathways. The line thickness represents the relative strength of the granger causality index of that pathway. The SN and thalamus were almost isolated from other nucleus and cortex. Bidirectional connection were identified between the right OFC and left NAc. (One-sample Wilcoxcon signed rank test, p<0.05, fdr corrected)
  • Simultaneous 3D 1H-MRSI and PET Imaging Associates Neurometabolism with Beta-amyloid Aggregation in Alzheimer's Disease
    Jialin Hu1, Miao Zhang2, Yaoyu Zhang1, Rong Guo3,4, Yudu Li3,4, Yibo Zhao3,4, Ziyu Meng1, Biao Li2, Jun Liu5, Binyin Li5, Jie Luo1, Chao Ma6, Georges El Fakhri6, Zhi-Pei Liang3,4, and Yao Li1
    1School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China, 2Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China, 3Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, United States, 4Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, United States, 5Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China, 6Gordon Center for Medical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
    We used simultaneous 3D MRSI and PET imaging to investigate association of neurometabolism with Aβ aggregation in HC, MCI and AD patients. Increase in mI and decrease in NAA were found as dementia severity increased.
    Figure 1. High-resolution metabolite maps and PET images simultaneously acquired from a HC, an MCI patient, and an AD patient, respectively. A global NAA reduction and mI elevation were observed with the increased dementia severity.
    Figure 3. Comparisons of neurometabolic concentrations among the HC, MCI, and AD groups for the global composite regions (top), PCC/precuneus (middle) and hippocampus (bottom). * p<0.05, ** p<0.01, *** p<0.001.
  • Non-invasive assessment of glycolytic and oxidative metabolism in mouse glioma using DGE 2H-MRS
    Rui Vasco Simoes1, Rafael N Henriques1, Beatriz M Cardoso1, Francisca F Fernandes1, Jonas L Olesen2, Sune N Jespersen2, and Noam Shemesh1
    1Champalimaud Research, Champalimaud Foundation, Lisbon, Portugal, 2Center of Functionally Integrative Neuroscience (CFIN) and MINDLab, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
    Glycolytic and oxidative turnover rates of glucose can be measured in mouse gliomas with DGE-2H-MRS. MP-PCA denoising improves the time-course detection and quantification of glucose oxidation, which in turn demonstrates correlation with MRI features of heterogeneity in the tumor region.
    Figure 2 – Improvement of DGE 2H-MRS spectral quality upon MP-PCA denoising. GL7: (A) T2-RARE showing the tumor region selected for DGE 2H-MRS (yellow dashed line); (B) SNRDHOi before and after denoising (average±SD); (C) left – eigenvalue spectrum from PCA decomposition (blue) and fit of MP distribution (orange), right – Gaussian distribution of denoising residuals, verified by the linearity of their logarithm; (D) DGE 2H-MRS data stacked, original and denoised, and spectral fitting including individual components, raw data, estimate and residual. * p=0.0003, paired t-Test.
    Figure 3 – Robustness of the kinetic model for different tumors and improvement of Glx fitting with MP-PCA denoising. Tumor volumes selected for DGE 2H-MRS (yellow dashed line) overlaid on reference T2-RARE transversal images for each animal (GL5-9, top). Fitting of Glc (red line), Glx (green line) and Lac (blue line) time-course changes displayed, before (center) and after denoising (bottom).
  • 1H-MRS of Primary Progressive and Relapsing-Remitting Multiple Sclerosis in brain white matter compared to healthy controls
    Bretta Russell-Schulz1, Jasmyne Kassam2, Michael Waine2, Erin L MacMillan1,3,4, Irene Vavasour1, Helen Cross2, Anthony Traboulsee2, Robert Carruthers2, and Shannon Kolind2,5,6
    1Radiology, UBC MRI Research Centre, Vancouver, BC, Canada, 2Medicine, University of British Columbia, Vancouver, BC, Canada, 3Philips Healthcare Canada, Markham, ON, Canada, 4Simon Fraser University's ImageTech Lab, Surrey, BC, Canada, 5Physics & Astronomy, University of British Columbia, Vancouver, BC, Canada, 6Radiology, University of British Columbia, Vancouver, BC, Canada
    This study establishes pre-treatment baseline metabolite concentrations for a longitudinal clinical trial for RRMS and PPMS using 1H-MRS. The high MRS data quality and similar in FWHM across all participants creates a strong baseline for detecting change over time.

    Figure 2a - Sample volume of interest (65x15x20mm3­) in white matter region on MPRAGE ‘anatomical’ scan for healthy control.

    b - Sample spectrum output from LCModel for each participant group with fit quality measures.

    Figure 4 - Tissue content within VOI separated by subject group and measures of spectrum fit quality (Full-Width-Half-Maximum, FWHM and Signal-to-Noise Ratio, SNR); FWHM and SNR. Where WMf, GMf, CSFf and Lesionf, are fraction of white matter, grey matter, cerebrospinal fluid and lesional tissue, respectively.
  • Evaluation of the agreement of metabolite levels between PRESS and MEGA-PRESS techniques in the grading of glioma patients
    Gerd Melkus1,2, Michael Taccone3,4, Ioana D Moldovan4,5, John Woulfe3,5,6, Gerard Jansen3,7, Ian Cameron1,2, Fahad AlKherayf3,4,5, and Thanh Binh Nguyen1,2
    1Medical Imaging, The Ottawa Hospital, Ottawa, ON, Canada, 2Radiology, University of Ottawa, Ottawa, ON, Canada, 3University of Ottawa, Ottawa, ON, Canada, 4Division of Neurosurgery, The Ottawa Hospital, Ottawa, ON, Canada, 5The Ottawa Hospital Research Institute, Ottawa, ON, Canada, 6Division of Neurology, University of Ottawa, Ottawa, ON, Canada, 7Department of Pathology and Laboratory Medicine, The Ottawa Hospital, Ottawa, ON, Canada
    T2 corrected metabolite levels of NAA, Cho and Cr obtained from the edit off spectrum of the MEGA-PRESS sequence are in good agreement with those obtained from a standard long TE PRESS sequence. No significant difference in the diagnostic accuracy was found.
    Figure 4. Diagnostic accuracy of the Cho/Cr ratio in the differentiation between high grade and low grade gliomas.
    Figure 1. Comparison of metabolite levels obtained from PRESS and edit-off spectrum using Bland-Altman analysis.
  • Quantification of neurobiological responses in the hippocampus: Towards in vivo neurochemical profiling of cuprizone-induced demyelination
    Do-Wan Lee1, Yeon Ji Chae2, Monica Young Choi2, Jae-Im Kwon3, Joongkee Min3, Chul‐Woong Woo3, Hwon Heo2, Dong‐Cheol Woo2,3, Jeong Kon Kim1, Kyung Won Kim1, Hyo Jeong Chin4, and Dong‐Hoon Lee4
    1Department of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea, Republic of, 2Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea, Republic of, 3Convergence Medicine Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Korea, Republic of, 4Department of Radiological Science, College of Health Sciences, Yonsei University, Wonju, Korea, Republic of
    The present study demonstrates how axonal demyelination affects cerebral metabolites in the gray matter of the hippocampal region, quantified using 7-T proton MR spectra.
    Figure 2. Representative in vivo 1H MR spectra in the right hippocampus of cuprizone-treated rats. The figure shows the raw spectrum (gray), fitted spectrum (navy), baseline (light blue), and 14 individual metabolite fits below (brown).
    Figure 3. Bar graph indicating the mean cerebral metabolite concentrations (a) and Cramer-Rao lower bounds (CRLBs) (b) in the right hippocampal region in control (CTRL) and cuprizone-treated (CPR) rats, quantified using the Linear Combination of Models software. The vertical lines on each of the bars indicate the (+) standard deviation of the mean values. *p < 0.05; **p < 0.01; ***p < 0.005.
  • Simultaneous Myelin Water Imaging and 3D 1H-MRSI Relates Myelin Degradation to Neurometabolic Changes in Mild Traumatic Brain Injury Patients
    Tianyao Wang1, Danni Wang2, Yujie Hu2, Rong Guo3,4, Yudu Li3,4, Yibo Zhao3,4, Jun Liu5, Zhi-Pei Liang3,4, and Yao Li2
    1Radiology Department, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China, 2School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China, 3Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, United States, 4Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, United States, 5Radiology Department, Tong Ren Hospital Shanghai Jiao Tong University School Medicine, Shanghai, China
    We investigated simultaneous myelin and neurometabolic alterations in acute mTBI patients. Our experimental results showed coupled myelin degradation and NAA reduction in the occipital corpus callosum. 
    Figure 1. Reconstructed MWF and neurometabolites maps of an acute mTBI patient and a healthy control. The neurometabolites include NAA, Cr, Cho and mI. The FOV covers the whole brain (240x240x120 mm3) and the data acquisition takes 8 minutes.
    Figure 2. A reduction in MWF was observed in the occipital tract of acute mTBI patients group and the spatially resolved spectra revealed a reduction of NAA and increase of mI in the patient.
  • Biochemical and behavioral alterations in a ferret model of blast related mild traumatic brain injury
    Shiyu Tang1,2, Su Xu1,2, Donna Wilder3, Joseph Long3, Venkata Siva Sai Sujith Sajja3,4, and Rao Gullapalli1,2
    1Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, United States, 2Center for Advanced Imaging Research (CAIR), University of Maryland School of Medicine, Baltimore, MD, United States, 3Blast Induced Neurotrauma Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States, 4The Geneva Foundation, Tacoma, WA, United States
    Metabolic changes using magnetic resonance spectroscopy and behavioral changes following blast injury were assessed. Metabolic changes in glutamate and taurine were observed concomitant increase in impulsivity at 1- and 3-months post blast TBI.
    Figure 1. A demonstration of the MRS voxel in prefrontal cortex and spectrums of a blast and sham ferret at 3-day post-blast.
    Figure 3. Glutamate and taurine levels in prefrontal cortex in sham and blast exposed ferret from 3-day to 3-month post-blast. *p<0.05, **p<0.01, group main effect; ##p<0.01, visit main effect.
  • Longitudinal Neurochemical Changes of Riluzole Therapy in Post-Traumatic Stress Disorder
    Sam H. Jiang1, David M. Benedek2, Patricia Spangler2, James West2, Catherine L. Dempsey2, Ashley Phares2, Brian Andrews-Shigaki3, Eduardo Coello1, and Alexander P. Lin1
    1Center for Clinical Spectroscopy, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States, 2Uniformed Services University of the Health Sciences, Bethesda, MD, United States, 3Office of Naval Research, United States Navy and Marine Corps, Alexandria, VA, United States
    The addition of riluzole to ongoing selective serotonin reuptake inhibitor therapy for combat-related post-traumatic stress disorder modulated glutamate-glutamine cycling, improved neural energetics, and induced cholinergic changes indicative of neuroprotective inflammation.
    Figure 3. Distributions of anterior cingulate cortex (ACC) a) phosphocholine + glycerophosphocholine (tCho) in the overall, riluzole, and placebo cohorts, and b) creatine + phosphocreatine (tCr) in the riluzole and placebo cohorts (* = p < 0.05, ** = p < 0.01).
    Figure 4. Distributions of amygdala a) glutamine (Gln) in the overall and placebo cohorts, and b) glutamate + glutamine (Glx) in the riluzole cohort (* = p < 0.05).
  • Temporal correlation of functional connectivity and Choline in the monkey brain following ischemic stroke
    Chun-Xia Li1, Frank Tong2, Doty Kempf1, Leonard Howell1, and Xiaodong Zhang1
    1Yerkes Imaging Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States, 2Department of Radiology, Emory University, Atlanta, GA, United States
    Previous studies have suggested cerebral Choline (Cho) is a sensitive marker of acute stroke and could protect the tissue from ischemic injury. Also the relative connectivity (RelCon) could be a robust index to reveal the functional connectivity changes using resting state fMRI (rs-fMRI). The results indicated progressively increased RelCon in secondary somatosensory cortex (RelCon-S2) and a significant positive correlation between RelCon-S2 and relative cerebral Choline level (RelCho) from hyper-acute phase to 96 hours post stroke. The RelCon and RelCho combined detection might be an optimized and promising approach in management and prediction of stroke recovery.
    Figure 1. A) The diffusion-weighted images of a stroke monkey brain demonstrated infarct evolution on Day 0, 2, and 4 post stroke (left). B) in vivo proton MR spectra in the contralateral and ipsilateral voxels in the stroke monkey brain 2 days post stroke. V1, MR Spectrum of contralateral voxel; V2, MR spectrum of ipsilateral area after MCA occlusion.
    Figure 2. A) Illustration of the representative slices of a stroke monkey brain with regions of interest of contralateral secondary somatosensory cortex (S2)(S2-CON, seed for functional connectivity analysis) and the ipsilateral S2 (S2-Ipsi). B) representative correlation map of S2 in a stroke monkey brain before stroke surgery (Pre) and post surgery. p = 0.026 with 20 voxels as threshold.
  • Standard Frame of Reference for the Identification of Metabolic Phenotypes of Brain Tumors using Single Voxel MR Spectroscopy
    Eduardo Coello1, Victoria Sanchez1, Marcia Louis1, Huijun Liao1, Sam Jiang1, Wufan Zhao1, Katherine M. Breedlove1, Raymond Huang1, and Alexander Lin1
    1Radiology, Brigham and Women's Hospital, Boston, MA, United States
    A method for the robust classification of MRS samples was developed. The model was able to correctly classify samples between tumor vs. non-tumor voxels, and IDH vs. non-IDH tumors.
    Fig. 2. Distribution of the quantified metabolite ratios in the analyzed datasets. The plot shows the variability of tumor tissue samples in comparison to the normal-appearing tissue.
    Fig. 4. Decision tree for the classification of tumor samples in normal-appearing tissue (normal) and tumor tissue.
  • GABA and Susceptibility Changes in Striatum in Liver Cirrhosis: Preliminary Results
    Gasper Zupan1,2, Sebastian Stefanovic3, Marjana Turk Jerovsek3, Borut Stabuc3, Georg Oeltzschner4,5, Stefan Ropele6, Dusan Suput1, and Andrej Vovk1
    1Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia, 2Institute of Radiology, University Medical Center Ljubljana, Ljubljana, Slovenia, 3Department of Gastroenterology and Hepatology, University Medical Center Ljubljana, Ljubljana, Slovenia, 4Russell H. Morgan Department of Radiology and Radiological Sciences, The John Hopkins University School of Medicine, Baltimore, MD, United States, 5M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, United States, 6Department of Neurology, Neuroimaging Research Unit, Medical University of Graz, Graz, Austria
    Liver cirrhosis in a systemic disease that affects brain as well. Using advanced MR methods, we demonstrated decreased striatal GABA levels, decreased susceptibility in caudate nucleus and increased susceptibility in putamen in patients with liver cirrhosis compared to healthy controls.
    Figure 4: Exemplary T1-, T2-weighted MR images and corresponding QSM (Quantitative Susceptibility Maps) of a LC patient (upper row) and a healthy control (lower row).
    Figure 2: Exemplary spectra of two LC pacients and a healthy control.
  • Upper brainstem GABA levels in Parkinson’s disease
    Yulu Song1, Tao Gong1, Muhammad G. Saleh2,3, Mark Mikkelsen2,3, Guangbin Wang 1, and Richard Edden2,3
    1Shandong Medical Imaging Research Institute, Shandong University, jinan, China, 2Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, baltimore, MD, United States, 3FM Kirby Center for Functional Brain Imaging, Kennedy Krieger Institute, baltimore, MD, United States
    We confirmed the hypothesis that a significant reduction in the GABA+ levels in the upper brainstem regions of patients with PD compared with the HCs.
    Bar Charts of the distributions of GABA+ levels, normalized fitting errors, linewidth in Hz, NAA, Cr, Cho levels PD Parkinson’s disease, HC healthy control, NAA N-acetyl aspartate, Cr creatine, Cho choline
    GABA+-edited spectra in the upper brainstem of all 36 participants, showing the intended signal at 3 ppm
  • In vivo detection of GSH in the mouse brain using MEGA-PRESS at 9.4T
    David Jing Ma1, Sabrina Gjerswold-Selleck2, Yanping Sun3, Matt Mattingly Mattingly4, and Jia Guo5
    1Biomedical Engineering, Columbia University, New York, NY, United States, 2Columbia University, New York, NY, United States, 3Herbert Irving Comprehensive Cancer Centre, Columbia University, New York, NY, United States, 4Bruker BioSpin, Billerica, MA, United States, 5Department of Psychiatry, Columbia University, New York, NY, United States
    MEGA-PRESS is a feasible technique to measure GSH in the mouse brain in vivo at 9.4T.
    Figure 4: Graphical representation of the GSH content in different brain regions. [A] Bar graphs of GSH/NAA at the thalamus and at the lateral ventricle with Student's t‐test result. It can be seen that there is a significantly less GSH/NAA content in the region around the lateral ventricle compared to the thalamus. [B] Bar graphs of GSH/NAA for the thalamus and lateral ventricular for each individual mouse. It can be seen that thalamus has significantly greater GSH/NAA than the lateral ventricle.
    Figure 2: GSH detection in the phantom solution with J-edited 1H MRS. Figure indicates single-voxel spectra of the 30 mM GSH phosphate buffer solution (PBS) acquired at 37 ºC in 4 min. [A] ‘ON’ Spectra with editing pulse applied at 4.56 ppm. [B] ‘OFF’ spectra with editing pulse applied at 8 ppm. [C] Difference spectra between [A] and [B] showing edited GSH resonance at 2.98 ppm. [D] GSH peak graphs at the following TE: 68, 90, 110, 130 and 150. [E] Plot of the GSH peak area under the curve versus TE.
  • Reproducibility of metabolite measurements in the preterm brain using magnetic resonance spectroscopy
    Subechhya Pradhan1,2,3, Sudeepta Basu4, Kushal Kapse1, Devon Fisher1, Stephanie Norman1, and Catherine Limperopoulos1,2,3
    1Developing Brain Institute, Children's National Hospital, Washington, DC, United States, 2Radiology, Pediatrics, George Washinton University, Washington, DC, United States, 3Radiology and Diagnostic Imaging, Children's National Hospital, Washington, DC, United States, 4Neonatalogy, Children's National Hospital, Washington, DC, United States
    Evaluation of reproducibility of metabolite measurements in the preterm infants using 4 MRS pulse sequences/ parameters showed good reproducibility for the largest number of metabolites using MEGA-PRESS sequence at TE = 68 ms and editing pulses at 1.9 and 7.8 ppm.
    Figure 2. Bar graph showing coefficient of variations for different metabolites measurements made using different pulse sequences
    Figure 1. Representative spectra showing spectra acquired from right basal ganglia using different pulse sequences.
  • Quantitative measurement of changes in 23Na MRI following transcranial direct current stimulation (tDCS) of the motor cortex
    Iris Asllani1,2, Francesco Di Lorenzo1, Balazs Orzsik1, Guillaume Madelin3, Neil Harrison4, and Mara Cercignani1
    1University of Sussex, Brighton, United Kingdom, 2Rochester Institute of Technology, Rochester, NY, United States, 3New York University, New York, NY, United States, 4University of Cardiff, Cardiff, United Kingdom
    • Anodal tDCS of M1 was associated with an increase in gray matter sodium concentration [Na]GM in the stimulated area measured using partial volume corrected 23MRI.
    • Subjects exhibited a widespread pattern of increased [Na]GM.
    • There were no changes in [Na]GM associated with sham tDCS.
    •  
    Fig.3: Areas in GM that survived the Δ%[NA] > 10% are shown for 2 randomly selected subjects. The arrows point to the "blob" that was identified under the marker on the MPRAGE (as demonstrated in Fig.1). The widespread pattern of Δ%[NA] was similar across the group.
    Fig.1: The motor ROI was selected by overlapping the GM Δ%[NA] image onto the subject's down-sampled MPRAGE. A MatLab script was written to identify "blobs" where Δ%[NA] > 10%. The script yields a blob ID number which is then used to extract the motor ROI for further analyses. Note the spatial relationship between the marker and the ROI.
  • Non-Invasive Brain Metabolic and Cytometric Imaging: Insights from Activity MRI [aMRI]
    Charles S. Springer1, Brendan Moloney1, Eric Baker1, Martin M. Pike1, and Xin Li1
    1Advanced Imaging Research Center, Oregon Health & Science University, Portland, OR, United States
    A map of awake, resting human brain homeostatic metabolic activity is shown for the first time.  Sodium Pump enzymatic turnover [fmol(ATP)consumed/s/cell] is particularly high in brain white matter.  This may have significant implications.
    Figure 1. The first aMRI maps of an axial image slice of the awake, healthy resting human brain. Panel (a) is the T1-weighted image. Panel (b) maps the cell density, r (cells/μL). Panel (c) maps the average cell volume, V (pL). Panel (d) maps the cellular water efflux rate constant, kio (s-1), reflecting cytoplasmic Sodium Pump enzymatic turnover. Note the particularly large kio values in white matter [panel (d)].