Arterial Spin Labelling
Diffusion/Perfusion Wednesday, 19 May 2021
Oral

Oral Session - Arterial Spin Labelling
Diffusion/Perfusion
Wednesday, 19 May 2021 12:00 - 14:00
  • Layer-dependent 7T ASL reveals sensory input and motor output perfusion activity in human primary motor cortex
    Xingfeng Shao1, Fanhua Guo2, Qinyang Shou1, Kai Wang1, Lirong Yan1,3, Kay Jann1,3, Peng Zhang2, and Danny JJ Wang1,3
    1Laboratory of FMRI Technology (LOFT), Mark & Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States, 2State Key Laboratory of Brain and Cognitive Science, Beijing MRI Center for Brain Research, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China, 3Department of Neurology, University of Southern California, Los Angeles, CA, United States
    Finger tapping (FT)-induced CBF increase shows a clear ‘double-peak’ pattern (somatosensory input in the superficial layers and motor output in the deep layers). Finger brushing (FB)-induced CBF increase was overall smaller, and mainly peaked in superficial layers (somatosensory input).
    Figure 4. (a) CBF map at rest (top) and two activation tasks (FT, middle; FB, bottom). (b) CBF increase evoked by the FT and FB tasks are highlighted in two insets. (c) Laminar profile of CBF at three tasks. (d) Laminar profile of CBF increase evoked by FT and FB. FT induced CBF increase shows a ‘double-peak’ pattern which corresponds to sensory input (superficial) and motor output (deep) layers respectively. FT induced CBF increase shows a weaker increase in superficial layers which corresponds to exteroception sensory input and minimal motor output.
    Figure 5. Six axial slices of the control images (a), resting-state perfusion maps (PLD=1000ms) (b) and CBF maps at rest, two activation tasks and the difference between rest and activation (blue boxes) (c). The proposed technique has sufficient coverage for perfusion measurement in M1 and supplementary motor area (SMA), etc.
  • Joint Estimation and Correction of Motion and Geometric Distortion in Segmented 3D Arterial Spin Labeling
    Jörn Huber1, Daniel Hoinkiss1, and Matthias Günther1,2
    1Fraunhofer MEVIS, Bremen, Germany, 2University of Bremen, Bremen, Germany
    Arterial Spin Labeling (ASL) yields functional information about the brain without exogenous contrast agents but ASL is sensitive to subject motion. In this work, a novel reconstruction for 3D GRASE PROPELLER ASL is presented, which jointly estimates motion and geometric distortion. 
    Figure 3: Reconstructed perfusion-weighted images with intentional motion and without intentional motion. The acquisition technique is indicated by 3D GRASE/3DGP and reconstruction without motion correction (NO), 3D GRASE three-dimensional rigid body motion correction (MOCO), standard PROPELLER motion correction (STD) and the proposed joint estimation of motion and distortion (JET) is applied. Note that in contrast to the original 3DGP method, a single motion estimate is calculated for each brick instead of the slice-by-slice approach to increase the robustness.

    Figure 4: a.) Procedure of 3DGP-JET. All bricks (three out of twenty control bricks from the third subject shown here) are used to estimate the underlying distortion field and individual motion parameters (Eq. 3). The distoriton field is then used to compensate distortion in individual bricks by unwarping with interpolated inverse field maps. Arrows indicate areas with large off-resonances and visible distortion in uncorrected 3DGP images; and b.) Mean and standard deviation of calculated motion parameters for the datasets without intentional motion using 3DGP-JET and 3DGP-STD.

  • Regulating labeling efficiency in arterial spin labeling using a multi-coil B0 shim array: Application to territory mapping
    Lincoln Craven-Brightman1, Yulin Chang2, Thomas Witzel3, Nicolas S. Arango4, Meher R. Juttukonda1,5, Luis Hernandez-Garcia6, Marta Vidorreta7, John A. Detre8,9, Lawrence L. Wald5,10, and Jason Stockmann5,10
    1Massachusetts General Hospital, Charlestown, MA, United States, 2Siemens Medical Solutions USA, Inc., Malvern, PA, United States, 3Qbio Inc, San Carlos, CA, United States, 4Dept. of Electrical Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States, 5Harvard Medical School, Boston, MA, United States, 6Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States, 7Siemens, S.A., Madrid, Spain, 8Neurology, University of Pennsylvania, Philadelphia, PA, United States, 9Radiology, University of Pennsylvania, Philadelphia, PA, United States, 10A.A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, United States
    We apply local B0 field control with a multi-coil (MC) shim array to improve ASL labeling. Labeling efficiency can be regulated by dynamically shimming the labeling plane. We demonstrate this capability through territory mapping by shifting target arteries out of the labeling pulse bandwidth.
    Figure 3. Experimental territory mapping using B0 local field control to selectively label internal carotid (IC) arteries. (a) The 1-cm labeling plane is shown with the baseline B0 field maps and voxel freq. histogram inside vertebral and IC artery masks. (b) Shimmed B0 maps and histograms showing spectral separation of the “desired” left IC from the “reject”. (c) Similar for right IC. Mean perfusion maps (difference between labeled and control case) at right show the applied shim field successfully shifts most of the “reject” arterial spins outside the passband of the labeling pulse.
    Figure 1. Loop geometry and photo/rendering of experimental and simulated B0 shim array hardware considered for ASL applications. B0 field map coverage for each array is shown at left. Representative element field maps are shown for both designs at level of slice shown, in the ASL labeling region. (a) 32-ch “AC/DC” shim array used for proof-of-concept territory mapping experiments. (b) Simulated 12-ch shim array specialized for ASL shimming applications, now under development.
  • Quantification of blood-brain barrier water permeability and arterial blood volume with multi-slice multi-delay diffusion-weighted ASL
    Hyun-Seo Ahn1, Jaeseok Park2, Chul Ho Sohn3, and Sung-Hong Park1
    1Department of Bio and Brain Engineering, Korea Advnaced Institute of Science and Technology, Daejeon, Korea, Republic of, 2Department of Biomedical Engineering, Sungkyunkwan University, Suwon, Korea, Republic of, 3Department of Radiology, Seoul National University Hospital, Seoul, Korea, Republic of
    • 1. The proposed multi-slice multi-delay diffusion-weighted ASL provides various quantitative perfusion measures including BBB permeability.
    • 2. The water exchange rate in Alzheimer patients were smaller than normal subjects, opposed to common knowledges on BBB permeability.
    Fig 3. From top to bottom, cerebral blood flow, arterial transit time, water exchange rate, arterial blood volume, and water permeability maps were estimated for 8 slices.
    Fig 2. (a) Sequence diagram of proposed 2D multi-slice multi-delay DWASL. (b) Bipolar diffusion weighting gradient was applied after slice excitation RF pulse which increased TE by 30ms. (c) Slice acquisition order was permuted through sets. 8 pairs of control and labeled image were obtained for each set. Slices of first half were imaged at odd PLDs, and second half were imaged at even PLDs.
  • phMRI with Simultaneous Measurement of Cerebral Perfusion and Blood-Cerebrospinal Fluid Barrier Function using Interleaved Echo-Time ASL
    Charith Perera1, Jack Wells1, Ian Harrison1, David Thomas2,3,4, and Mark Lythgoe1
    1Centre for Advanced Biomedical Imaging, University College London, London, United Kingdom, 2Neuroradiological Academic Unit, Department of Brain Repair and Rehabilitation, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom, 3Leonard Wolfson Experimental Neurology Centre, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom, 4Wellcome Centre for Human Neuroimaging, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
    CO2 induced similar increases in cortical perfusion and the BCSFB-ASL signal (21%). Caffeine evoked an exclusive, marked decrease in BCSFB-mediated water exchange (41%). The aged brain displays impaired BCSFB reactivity towards a vasopressin challenge.
    Figure 1 - Interleaved-TE ASL data obtained simultaneously for relative cortical perfusion and relative BCSFB-ASL signal. Averaged time courses for relative cortical perfusion (column 1) and the individual animal averages (column 2). Relative BCSFB-ASL signal averaged time courses (column 2) are shown alongside their respective individual animal averages (column 3). Rows represent each drug or challenge: row 1 - CO2 (a-d), row 2 - vasopressin (e-h), row 3 - caffeine (i-l). Further details of the drug challenges can be found in Table 1.

    Figure 2 - Adult (n = 14) vs aged (n = 14) response to vasopressin.

    a) Relative BCSFB-ASL signal obtained using Interleaved-TE ASL, showing both adult and aged averaged time courses (vasopressin administration at 10 mins).

    b) Adult (n = 14) vs aged (n = 14) response to vasopressin, individual animal averages (baseline and post-vasopressin challenge).

  • Venous Oxygenation Mapping using Fourier-Transform based Velocity-Selective Pulse Trains
    Wenbo Li1,2, Peter van Zijl1,2, and Qin Qin1,2
    1Radiology Department, Johns Hopkins University School of Medicine, Baltimore, MD, United States, 2Kirby Image Center, Kennedy Krieger Institute, Baltimore, MD, United States
    A T2-oximetry technique for mapping venous oxygenation based on Fourier-transform based velocity-selective pulse trains is proposed with the advantage of higher SNR. Preliminary data on two healthy subjects show that the averaged Yv across all the voxels is comparable to the global Yv.
    Figure 3. The DIR (GM) map, S0 map, T2v (ms) map and Yv maps for the two subjects. The extrapolated S0 values are proportional to the venous CBV and, as expected, show higher signal in the GM than in the WM. The fitted T2 maps and the corresponding Yv maps reveal a more uniform contrast between GM and WM, as expected for brain OEF. Several voxels (mainly in white matter region) with very low signal intensities were excluded during the data processing.
    Figure 2. a) Idealized cartoon depicting the evolution of the longitudinal magnetization of water in different compartments. Relaxed and dephased spins are denoted by upright arrows and solid circles, respectively. b) Simulated magnetization evolution for the arterial and venous blood. The consecutive FT-VSI and NSI pulses invert spins flowing above the VCUTOFF for arterial nulling and preserve the spins moving below the VCUTOFF. These preserved spins will largely outflow into the venules during TI, thus yielding a high SNR.
  • VESPA ASL: VElocity and SPAtially selective Arterial Spin Labeling
    Joseph G Woods1, Eric C Wong1, Emma Boyd2, and Divya S Bolar1
    1Radiology, UCSD, La Jolla, CA, United States, 2Neurosciences, UCSD, La Jolla, CA, United States
    VESPA ASL is introduced, which simultaneously acquires VSASL and PCASL data. A signal model to quantify cerebral blood flow and arterial transit times (ATT) from these data is presented, with generation of quantitative CBF and ATT maps in vivo. 
    Figure 3: The VESPA decoded VSASL and PCASL data, with the CBF and ATT maps calculated by voxelwise fitting the VESPA signal model to the decoded VESPA data. Five slices from 1 subject are shown. Longer ATTs can be seen in the white matter, posterior circulation, and at both internal and external border zones. Before fitting, the data were voxelwise calibrated by the M0 image and corrected for slice-dependent T1 decay.
    Figure 1: Sequence diagrams, label/control encoding, and contrast decoding strategies for PCASL, VSASL and VESPA. In VESPA, VSASL and PCASL are combined and used to label different pools of inflowing arterial blood. VSASL labeling (VS1) uses a spatially-selective VSASL module surrounding the imaging slab. The PCASL labeling plane is immediately inferior to the VSASL slab. By alternating label and control conditions in different orders, VSASL and PCASL contrasts can be linearly encoded into the acquired images and decoded by simple image arithmetic
  • Faster regional cerebral blood flow increases in infant heteromodal cortex with 2.5mm3 resolution 3D multi-shot, stack-of-spirals pCASL
    Minhui Ouyang1, John Detre2, Chenying Zhao1,3, Samantha Lam1, J. Christopher Edgar1,2, and Hao Huang1,2
    1Department of Radiology, The Children's Hospital of Philadelphia, Philadelphia, PA, United States, 2Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States, 3Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States
    Heterogenous maturational pattern of regional cerebral blood flow in infants aged 0-18months, with maturation most prominent in heteromodal association cortex, was revealed by using a high-resolution 3D multi-shot, stack-of-spirals pCASL perfusion MRI at isotropic 2.5mm.
    Fig. 2: High-resolution (2.5x2.5x2.5mm3) regional cerebral blood flow (rCBF) maps acquired with 3D multi-shot, stack-of-spirals pCASL, from three representative infants aged 2months (A), 8months (B) and 17months (C).
    Fig. 3: Main effect of age on cerebral blood flow (CBF) during infancy. (A) Global CBF increases during infancy. Data points represent global CBF measured with phase-contrast MRI of each subject. (B) rCBF increases throughout the cortex, but more prominently in the heteromodal association cortex. Images thresholded at t(df=21) > 2.83 with p<0.01. (C) heterogeneous developmental rate of rCBF (ml/100g/min/month) across cortex during infancy. Slower and faster rCBF growth rates are shown in cool and warm colors, respectively.
  • Spatiotemporal characteristics of longitudinal changes in cerebral blood flow across the adult lifespan
    Hualu Han1,2, Zixuan Lin2,3, Melissa Rundle4, Anja Soldan5, Corinne Pettigrew5, Joshua F. Betz6, Kumiko Oishi7, Yang Li2, Binu P. Thomas8, Peiying Liu2, Marilyn Albert5, Denise Park4, and Hanzhang Lu2,3,9
    1Center for Biomedical Imaging Research, Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, China, 2The Russell H. Morgan Department of Radiology & Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States, 3Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States, 4Center for Vital Longevity, School of Behavioral and Brain Sciences, University of Texas at Dallas, Dallas, TX, United States, 5Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States, 6Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States, 7Center for Imaging Science, Johns Hopkins University, Whiting School of Engineering, Baltimore, MD, United States, 8Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX, United States, 9F.M. Kirby Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, United States
    Longitudinal CBF decrease with age and the longitudinal rate of decline was faster than that from the cross-sectional data. CBF reduction is faster in younger than in older individuals and has spatial differences and hemispheric asymmetry.
    Figure 1. Regional CBF (mean ± standard error) between baseline and follow-up in major GM structures of Study 2.
    Table 3. Linear mixed models for assessing the changes of global CBF and normalized volume in Study 1 (N=127) and Study 2 (N=115). Age, time interval, gender, GM probability and WM probability were added as the fixed effects in Model 1, and age-by-follow-up time interaction was additionally added in Model 2.
  • Quantification of arterial obstruction in pediatric patients with pulmonary embolism using arterial spin labeled perfusion MRI of the lungs
    Joshua S Greer1,2,3, Mubeena Abdulkarim1, Gerald F Greil1,3, Ayesha Zia1, Ananth J Madhuranthakam2,3, and Tarique Hussain1,2
    1Pediatrics, UT Southwestern Medical Center, Dallas, TX, United States, 2Radiology, UT Southwestern Medical Center, Dallas, TX, United States, 3Advanced Imaging Research Center, UT Southwestern Medical Center, Dallas, TX, United States
    Perfusion imaging using ASL was demonstrated in pediatric patients with pulmonary embolism, which successfully detected perfusion defects in all patients. A method to quantify pulmonary vascular obstruction was developed using ASL, which showed moderate agreement with CT angiography.
    Figure 3: (A) MS-FAIR perfusion and CTA images in a patient with extensive PE at diagnosis. Red arrows show CTA pulmonary artery filling defects, which correspond well with ASL perfusion defects. Yellow arrows indicate vessels with normal filling on CT that correspond well with regions of higher ASL perfusion. (B) The same patient 6 months after tPA treatment, showing significantly improved perfusion. PVO before and after treatment were 73% and 8% measured by CT, and 63% and 39% by ASL. A residual defect was detected by ASL (B, red arrows), while the vascular occlusion by CT had resolved.
    Figure 1: (A) Sagittal MS-FAIR quantified perfusion images in a patient with extensive filling defects across the right lung noted by CTA 6 days prior to ASL imaging. The edges of the right lung are outlined in red. Average PVO for this patient was 55% measured by CTA, and 44% measured by ASL. (B) Overlaid PVO values on the perfusion images, with higher degrees of obstruction corresponding well with the perfusion defects in Fig 1A. Overlaid PVO values under 50% were made transparent for visualization purposes
Back to Top
Digital Poster Session - Arterial Spin Labelling: Methods
Diffusion/Perfusion
Wednesday, 19 May 2021 13:00 - 14:00
  • The Open Source Initiative for Perfusion Imaging (OSIPI): ASL Pipeline inventory
    Sudipto Dolui1, Hongli Fan2, Paula L. Croal3,4, Charlotte Buchanan4, Lydiane Hirschler5, Udunna Anazodo6, David L. Thomas7, Henk J.M.M. Mutsaerts8, and Jan Petr9
    1Department of Radiology, University of Pennsylvania, Philadelphia, PA, United States, 2The Johns Hopkins School of Medicine, Baltimore, BC, United States, 3Radiological Sciences, Division of Clinical Neuroscience, School of Medicine, University of Nottingham, Nottingham, United Kingdom, 4Sir Peter Mansfield Imaging Centre, School of Medicine, University of Nottingham, Nottingham, United Kingdom, 5Leiden University Medical Center, Leiden, Netherlands, 6Western University, London, ON, Canada, 7UCL Queen Square Institute of Neurology, University College London, London, United Kingdom, 8Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, location VU, Amsterdam, Netherlands, 9Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
    As a part of the Open Source Initiative for Perfusion Imaging (OSIPI), we have created an inventory of pipelines for processing of arterial spin labeling data. We listed their features and software requirements to facilitate easy access based on user’s requirements.
    Table 2. Requirements. This table provides the basic requirements on the operating system, software, and data input types. It further lists the compatible vendors (note that other vendors might also work on each pipeline even though they were not tested for it). All pipelines were developed and tested to work with human brain scans. The last column lists other organs and animals that were tested.
    Table 1. Contact details. This table contains the basic details about the pipeline such as the pipeline author, affiliation, and a link to a scientific publication with more details about the pipeline. Furthermore, it is indicated if the pipeline is available on request or by direct download (see TF 1.1 website15 for the links to the pipelines and author contacts). Last columns indicate if the pipeline is provided with a manual or guide, open source code, and a graphical user interface (GUI).
  • The Open Source Initiative for Perfusion Imaging (OSIPI) ASL MRI Challenge
    Udunna Anazodo1,2, Joana Pinto3, Flora A. Kennedy McConnell4,5,6, Maria-Eleni Dounavi7, Cassandra Gould van Praag8, Henk Mutsaerts9, Aaron Oliver Taylor10, André Paschoal11, Jan Petr12, Diego Pineda-Ordóñez13, Joseph G. Woods14, Moss Y. Zhao15, and Paula L. Croal4,5
    1Department of Medical Biophysics, University of Western Ontario, London, ON, Canada, 2Imaging Division, Lawson Health Research Institute, London, ON, Canada, 3Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, United Kingdom, 4Sir Peter Mansfield Imaging Centre, University of Nottingham, Nottingham, United Kingdom, 5Radiological Sciences, Division of Clinical Neuroscience, School of Medicine, University of Nottingham, Nottingham, United Kingdom, 6Nottingham Biomedical Research Centre, Queens Medical Centre, Nottingham, United Kingdom, 7Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom, 8Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, United Kingdom, 9Amsterdam University Medical Center, Amsterdam, Netherlands, 10Gold Standard Phantoms Limited, London, United Kingdom, 11Department of Radiology, LIM44 - HCFMUSP, Sao Paulo, Brazil, 12Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany, 13Department of Radiology, Clinica Del Country, Bogotá, Colombia, 14Department of Radiology, University of California San Diego, San Diego, CA, United States, 15Department of Radiology, Stanford University, Stanford, CA, United States
    The OSIPI ASL MRI Challenge aims to establish the range of image analysis approaches used for perfusion quantification and identify optimum pipelines, ultimately moving towards community consensus for reproducible analysis of ASL MRI.
    Figure 1: Projected timeline for the OSIPI ASL Challenge. The challenge will be open for a period of six months, with winning teams presenting at the ISMRM Perfusion Study Group meeting at ISMRM 2022.
    Figure 2: Example perfusion-weighted images (arbitrary units) for A) population-based and B) synthetic datasets.
  • Optimization of post-labeling delays in multi-delay 3D pCASL by modeling arterial transit time distribution
    Zhiyuan Zhang1,2, Timothy Macaulay3, and Lirong Yan1
    1USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States, 2Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, United States, 3Division of Biokinesiology and Physical Therapy, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, United States
    Multi-delay ASL with PLDs optimized by modeling ATT as normal distribution provides more accurate CBF and ATT quantifications compared to that with evenly spaced PLDs or optimal PLDs based on uniform ATT distribution.
    Figure 2. a) Three ATT distribution profiles used for PLD optimization b) and generated optimal PLDs for each ATT distribution along with evenly spaced PLDs .
    Figure 3. The estimated CBF and ATT maps and corresponding error maps with the four ASL protocols at SNR of 15dB from a subject. Ground truth was CBF map and ATT map calculated from the in vivo ASL scan, respectively.
  • Investigation of angiographic shine-through in time-encoded pCASL
    Lena Vaclavu1, Dilek Betül Arslan2, Lydiane Hirschler1, Carles Falcon3,4, Esin Ozturk-Isik2, Juan Domingo Gispert3,4, Paula Montesinos5, Kim van de Ven6, and Matthias JP van Osch1
    1Department of Radiology, C.J. Gorter Center for High Field MRI, Leiden University Medical Center, Leiden, Netherlands, 2Biomedical Engineering Institute, Boğaziçi University, Istanbul, Turkey, 3BarcelonaBeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain, 4CIBER-BBN, Madrid, Spain, 5Philips Healthcare Iberia, Madrid, Spain, 6Philips Healthcare, Best, Netherlands
    Our results show that increased signal fluctuations in arterial vessels in time-encoded pCASL is associated with selective background suppression and is independent of the Hadamard labeling process.
    Figure 1. a) An example of the shine-through effect in a healthy volunteer seen in the perfusion block of a fully acquired time-encoded pCASL (te-pCASL) dataset noticeable as negative signal in the large arteries and/or positive signal in higher slices. b) The temporal standard deviation (tStdv) of the same dataset shows that the effect (high tStdv in vessels) is present in all post-label-delays (PLDs) and that tStdv is an insightful measure of the effect.
    Figure 4. ASL (subtracted) signal is shown for a single slice low in the brain, for the seven individual experiments in which RF (labeling) was enabled for single blocks only (columns). The separation of the individual blocks in each experiment can be seen in the black squares which have positive ASL signal. We observed that the shine-through artefact (red arrows) was visible in the disabled blocks, (negative or positive signal), and surprisingly, also in the experiment without any labeling at all (last column, experiment #8). See Figure 1 for explanation of the experiment numbers.
  • Verifying the effect of DANTE preparation pulse for separating spin-compartments in arterial spin labeling using T2-measurement
    Shota Ishida1, Hirohiko Kimura2, Naoyuki Takei3, Yasuhiro Fujiwara4, Tsuyoshi Matsuda5, Yuki Matta1, Masayuki Kanamoto1, Nobuyuki Kosaka2, and Eiji Kidoya1
    1Radiological center, University of Fukui Hospital, Eiheiji, Japan, 2Department of Radiology, Faculty of Medical Sciences, University of Fukui, Eiheiji, Japan, 3Global MR Applications and Workflow, GE Healthcare Japan, Hino, Japan, 4Department of Medical Image Sciences, Faculty of Life Sciences, Kumamoto University, Chuo-ku, Japan, 5Division of Ultra-high Field MRI, Institute for Biomedical Science, Iwate Medical University, Shiwa-gun, Japan
    The efficacy of DANTE for vascular suppression was validated by direct measurement of T2 values of ASL signals under the application of DANTE. DANTE separates the vascular and tissue compartments without image degradation.
    Averaged ASL images of each PLD with different eTEs and T2 maps in the MNI-space. The left and right panels show the acquired images without and with DANTE, respectively.
    Averaged CBF and transit time (TT) maps in the MNI-space.
  • Systematic investigation of the sensitivity of optimised pCASL protocols to macrovascular contamination
    Logan Xin Zhang1 and Michael A Chappell1,2,3
    1Institute of Biomedical Engineering, University of Oxford, Oxford, United Kingdom, 2Radiological Sciences, Division of Clinical Neurosciences, School of Medicine, University of Nottingham, Nottingham, United Kingdom, 3Sir Peter Mansfield Imaging Centre, School of Medicine, University of Nottingham, Nottingham, United Kingdom
    An extended kinetic model explicitly accounting for macrovascular signal is beneficial to CBF estimation in pCASL protocols, reducing CBF sensitivity to macrovascular contamination to up to 2.3 seconds of tissue ATT. 
    Figure 2. CBF sensitivity maps to macrovascular contamination by eight protocols (a to h) and average (i). Each protocol has CBF maps estimated by GKM (left), GKMmvc (middle), and their difference (GKMmvc - GKM, right). Units: error% per 1% aBV.
    Figure 3. CBF sensitivity as a function of tissue ATT across protocols under three assumptions. (a) Percentage; (b) Fixed; (c) Fixed interval. Each assumption has CBF maps estimated by GKM (left), GKMmvc (middle), and their difference (GKMmvc - GKM, right). Units: error% per 1% aBV.
  • Perfusion Quantification using Velocity Selective Inversion pulses in a combined ASL-MRF Framework
    Anish Lahiri1, Jeffrey Fessler1, and Luis Hernandez-Garcia2
    1Electrical and Computer Engineering, University of Michigan, Ann Arbor, MI, United States, 2FMRI Laboratory and Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
    A combination of Velocity Selective Inversion pulses and ASL-MRF may be a feasible alternative to pCASL-based ASL-MRF for quantification of perfusion and other hemodynamic parameters in the brain
    Figure 4: Estimates for Perfusion, BAT and tissue T1 obtained from a healthy human subject using our combined VSI and MRF-ASL framework.
    Figure 1: Composition of a single repetition time (TR) and details of the pulse sequence used in our work. The TR is varied across acquisitions by varying the Post Labeling Delays.
  • A general framework for eddy current minimization in Velocity Selective Arterial Spin Labeling
    Joseph G. Woods1, Eric C. Wong1, David D. Shin2, and Divya Bolar1
    1Department of Radiology, University of California San Diego, San Diego, CA, United States, 2GE Healthcare, Menlo Park, CA, United States
    A novel framework that minimizes eddy current artifacts in velocity-selective (VS) arterial spin labeling is introduced. The framework is applicable to any VS module design with no time penalty. Simulation and empirical data are presented with artifact reduction in phantom and human data.
    Phantom error maps (% error) reflecting residual eddy current effects before and after VS gradient optimization, for DHRS and BIR8 VS labeling schemes. The bar graph reflects the mean absolute error across the phantom volume. Significant improvements are seen for the x, y, and z gradient axes for DHRS. The BIR-8 errors are small for each gradient axis, and little or no improvement was seen.
    Human perfusion-weighted images acquired before and after VS gradient optimization for both DHRS and BIR-8 VS modules in x and y gradient directions. z direction was not acquired due to subject intolerance. Marked EC effects are seen for the standard DHRS approach, which are qualitatively improved after optimization. BIR-8 errors are small for each gradient axis, and little or no improvement was seen.
  • Reduction of motion effects in myocardial arterial spin labeling
    Verónica Aramendía-Vidaurreta1,2, Pedro M. Gordaliza3,4, Marta Vidorreta5, Rebeca Echeverría-Chasco1,2, Gorka Bastarrika1,2, Arrate Muñoz-Barrutia3,4, and María A. Fernández-Seara1,2
    1Radiology, Clínica Universidad de Navarra, Pamplona, Spain, 2IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain, 3Universidad Carlos III de Madrid, Madrid, Spain, 4Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain, 5Siemens Healthineers, Madrid, Spain
    Results indicate the superiority of acquiring myocardial ASL images using a free-breathing strategy combined with pairwise registration, showing higher accuracy (in synthetic images) and higher reproducibility together with lower variability across subjects (in in vivo images).
    Figure 4: Bland-Altman plots and within-subject coefficient of variation (wsCV) obtained from the global myocardial perfusion measurements in the in vivo datasets
    Figure 3: Boxplots of MBF measurements in vivo averaged across the two intrasession acquisitions for the different breathing strategies (BH=breathhold, SB=synchronized breathing and FB=free breathing) and post-processing steps (O=Original dataset, P= after pairwise registration, G= after groupwise registration, DD = after detecting and discarding outliers). *Indicates statistically significant differences between datasets (p=0.05).
  • Clinically applicable automatic quantitative renal perfusion measurement using ASL-MRI and machine learning
    Isabell Katrin Bones1, Clemens Bos1, Chrit Moonen1, Jeroen Hendrikse2, and Marijn van Stralen1
    1Center for Image Sciences, University Medical Center Utrecht, Utrecht, Netherlands, 2Department of Radiology, University Medical Center Utrecht, Utrecht, Netherlands
    Automatic and manual segmentations showed good agreement and delivered comparable cortical renal blood flow when used for ASL quantification. Thus, automatic segmentation using a cascade of U-nets makes renal ASL-MRI quantification feasible.
    Single slice segmentation example. Segmentations are displayed in blue contours. (A) M0-image with whole kidney contours as a result from U-Ne2. T1-map and perfusion map with cortical contours as a result from U-Net3, corrected with U-Net2 output. (B) Reference cortical contours manually drawn by observer1. (C) Cortical contours manually drawn by observer2. Good agreement between the three different cortical contours can be seen; the bright cortical perfusion signal is captured by all contours, assuring accurate mean cortical RBF calculation.
    Graphic representation of our segmentation cascade for renal cortical voxel extraction. U-net1 roughly localizes both kidneys on full FOV M0-images. This information is used for image cropping, to remove background which permits faster image registration. U-net2 segments fine whole kidney masks on the cropped and registered M0-images. U-net3 is predicting cortical voxels from cropped and registered T1-maps. In a last step, the predicted cortical and whole kidney masks from U-Net2 and U-Net3 are combined to remove eventual erroneous cortical predictions outside of the kidney.
  • Arterial Spin Labeling Denoising with Convolutional Neural Network and Convolutional Long-Short-Term-Memory (ConvLSTM)
    Qinyang Shou1, Chaitanya Gupte2, Danny JJ Wang1, and Hosung Kim2
    1Laboratory of Functional MRI Technology (LOFT), Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, United States, 2Neuroimaging with Deep Learning Lab (NIDLL), Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, United States
    Our model combining CNN with Convolutional LSTM for spatiotemporal ASL denoising can achieve an SNR improvement with fewer input time steps, which shows a potential to reduce the scan time for ASL acquisition.
    Figure1. Network structure of the denoising model. The network is consisted of a local pathway and a global pathway. The local pathway is combined with five layers of 3D convolutional layers and one 2D ConvLSTM layer and a 3D convolutional layer at the end. Inputs to the network are several perfusion image series and the outputs are the denoised perfusion image.
    Figure 3 Model Outputs with different input time steps. The predictions are produced with 20, 15, 10 and 5 time steps as input using the corresponding model.
  • Gradient adjustments for improved pcASL exploiting a B1+ shimmed labeling train
    Christian R. Meixner1, Sebastian Schmitter2,3, Jürgen Herrler4, Arnd Dörfler4, Michael Uder1, and Armin M. Nagel1,3
    1Institute of Radiology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany, 2Physikalisch-Technische Bundesanstalt (PTB), Braunschweig und Berlin, Germany, 3Division of Medical Physics in Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany, 4Institute of Neuro-Radiology, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
    With lower slice-selective gradient amplitude of the pcASL labeling – exploiting B1+-phase shim and Variable-Rate Selective Excitation – the temporal signal-to-noise ratio of perfusion images can be improved at 7T under suboptimal B1+ conditions.
    Figure 3: Simulation results: a) labeling efficiencies of the different slice selective gradients and B1+ amplitudes, as well as the mean B1+ amplitude of the 5 subjects after shimming and the required B1+ amplitude. b) normalized labeling slice profiles of the different slice selective gradients: with lower gradient amplitude the labeling slice gets thicker and vice versa.
    Figure 5: Example of one subject with 7mT/m and the individual adjusted shim.
  • Whole-brain Perfusion Mapping at 7T by SAR-efficient Non-segmented 3D EPI-pCASL
    Seon-Ha Hwang1, SoHyun Han2, Seong-Gi Kim2, Jaeseok Park3,4, and Sung-Hong Park1
    1Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea, Republic of, 2Center for Neuroscience Imaging Research, Institute of Basic Science, Suwon, Korea, Republic of, 3Department of Biomedical Engineering, Sungkyunkwan University, Suwon, Korea, Republic of, 4Department of Intelligent Precision Healthcare Convergence, Suwon, Korea, Republic of
    The proposed SAR-efficient non-segmented 3D-EPI-pCASL produced high-quality perfusion maps at both 3T and 7T. Its SAR efficiency and high temporal resolution enabled us to get whole-brain 3D-pCASL fMRI map at 7T for the first time.
    Fig. 3. Representative percent signal change map of 3D EPI-pCASL and spatial SNR of perfusion map at 7T. (a) CBF map by 3D EPI-pCASL with 5mm slice thickness. (b) Table of spatial SNR of perfusion map by thickness. Despite limitation of coil coverage and B0, B1 inhomogeneity, the perfusion map from 7T MRI have consistent quality with that of 3T. Also, spatial SNR is similar with that of 3T. Because lowered labeling efficiency by hardware limitation could not be calculated, percent signal change map is represented instead of CBF map.
    Fig. 4. Functional activation map by (a)perfusion and (b)BOLD activation. For the both perfusion and BOLD, activations were shown simultaneously at visual (occipital lobe) and motor cortex (white arrows). Although the sensitivity of perfusion activation is lower than that of BOLD, spatial localization is much specified by perfusion activation map.
  • PCASL labeling efficiency measurement with B0 off-resonance compensation at 7T
    Gael Saib1, Alan Koretsky1, and S Lalith Talagala2
    1NINDS/LFMI, National Institutes of Health, Bethesda, MD, United States, 2NINDS/NMRF, National Institutes of Health, Bethesda, MD, United States
    The maximal labeling efficiency of PCASL at 7T was experimentally measured at ~0.6 using a 1Tx/32Rx head coil with a PCASL-prepared FLASH sequence.
    Figure 2: PCASL labeling efficiency as a function of B1mean measured in ICAs with Gmean 0.25mT/m. For 4 of the arteries, the labeling efficiency maximized ~0.6 at a B1mean of ~0.4uT. Note that the maximum achievable B1mean was limited in RICA of subject 3 compared to others due to increased B1+ inhomogeneities in the labeling plane.
    Figure 4: Whole-brain CBF maps acquired with a 2D-EPI PCASL sequence at 7T. The PCASL sequence parameters were Gmax=3.5mT/m, Gmean=0.25mT/m, Hanning RF pulse duration/separation=800/1800s, nominal FA=35°, labeling duration/PLD=1.5s, matrix=64x64, resolution=3x3x3mm3, 23 slices, TE/TR=12/4500ms, GRAPPA factor=2 and TA=6min. With a labeling efficiency of 0.6, the average CBF in the cortex was about 66mL/100g/min.
  • Retrospective Motion Correction of multi-shot 3D GRASE Arterial Spin Labelling using ESPIRiT reconstruction
    Jack Highton1, Enrico De Vita2, and David Thomas3,4,5
    1UCL Queen Square Institute of Neurology, University College London, London, United Kingdom, 2School of Biomedical Engineering and Imaging Sciences, Kings College London, London, United Kingdom, 3Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom, 4Neuroradiological Academic Unit, Department of Brain Repair and Rehabilitation, University College London, London, United Kingdom, 5Wellcome Centre for Human Neuroimaging, University College London, London, United Kingdom
    A retrospective motion correction method for 3D GRASE ASL used a combination of ESPIRiT and image registration. Experimentally, it reduced artifacts and increased correlation between cerebral blood flow measurements acquired with and without motion.
    Figure 5 - Top: CBF map slices from the experiment, calculated from 10 ASL repeats with the following motion circumstance: a) No deliberate motion. b) Nodding motion. c) Nodding motion with ESPIRiT based motion correction. Bottom: d) Correlation between the partial volume corrected voxel GM measured when the test subject was deliberately nodding v.s. when they were remaining still. e) Equivalent correlation after ESPIRiT based motion correction was applied.
    Figure 2 - 1) The raw k-space data from all four shot acquisitions from all control image repeats are used to calculate coil sensitivity maps. 2) These are used to reconstruct full images from each of the individual shots. 3) These are then registered by maximizing mutual information. 4) The corrected tag and control image are registered by minimising the standard deviation of the subtraction, found to be the best metric for this problem. 5) The motion corrected data is used to calculate CBF maps.
  • Optimization and Evaluation of Super-Resolution SMS ASL with Slice-Dithered Enhanced Resolution (SLIDER) technique
    Qinyang Shou1, Xingfeng Shao1, and Danny JJ Wang1
    1Laboratory of Functional MRI Technology (LOFT), Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, United States
    We optimized and evaluated super-resolution perfusion imaging using SLIDER SMS ASL. A greater number of shifted slices is beneficial for SNR efficiency, at the cost of increased slice blurring.
    Figure 1: SLIDER acquisition scheme. (a) SMS-EPI pCASL acquisition sequence for each set of low-resolution images, with constrained slice dependent background suppression. (b)Acquisition scheme for SLIDER2, SLIDER3 and SLIDER4, different number of scans with slice shift are needed, 2 for SLIDER2, 3 for SLIDER3 and 4 for SLIDER4. Then high-resolution images can be reconstructed.
    Figure2: SLIDER super-resolution reconstructed ASL images and high-resolution ASL images in the axial direction. The reconstructed images well follow the high-resolution images, with an improved SNR. The green and red boxes and corresponding zoomed images show two typical slices with SNR improvement.
  • Saturated Look-Locker FAIR (SALL-FAIR) Sequence with FPOCK Model for Simultaneous Acquisition of CBF, aBAT, tBAT, and T1 Map
    Zihan Ning1, Shuo Chen1, Zhensen Chen1, Huiyu Qiao1, Hualu Han1, Rui Shen1, Dandan Yang1, and Xihai Zhao1
    1Center for Biomedical Imaging Research, Department of Biomedical Engineering, School of Medicine Tsinghua University, Beijing, China
    The proposed saturated Look-Locker FAIR (SALL-FAIR) sequence with FPOCK model could provide accurate CBF, aBAT, tBAT, T1 map with a single scan.
    Figure 3. (A) Perfusion weighted signal ΔM [% of M0] from Subject 3 from TI = 250-1650 ms; (B) T1 map (ms) (C) CBF, aBAT tBAT and RMSE maps using the single-TI model, the Buxton’s model and FPOCK model.
    Figure 1. Pulse sequence diagram for the SALL-FAIR sequence (A) and the illustration of relative spatial positions of RF pulses (B).
  • BATS: the Boston ASL Template and Simulator – development and initial evaluation
    Manuel Taso1, Fanny Munsch1, and David C Alsop1
    1Division of MRI research, Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
    We  report here the initial development of BATS – the Boston ASL Template and Simulator, highlighting its development and initial use as a group-average, as a registration target for individual scans and as a synthetic ASL contrast generator. 
    Figure 1 – Anatomical and perfusion templates included in BATS
    Figure 5 – Simulated ASL signal at different PLDs
  • ASLDRO: Digital reference object software for Arterial Spin Labelling
    Aaron Oliver-Taylor1, Thomas Hampshire1, Nadia A S Smith2, Michael Stritt3, Jan Petr4, Johannes Gregori3, Matthias Günther3,5, Henk J Mutsaerts6, and Xavier Golay1,7
    1Gold Standard Phantoms Limited, London, United Kingdom, 2National Physical Laboratory, Teddington, United Kingdom, 3mediri GmbH, Heidelberg, Germany, 4Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany, 5Fraunhofer MEVIS, Bremen, Germany, 6Amsterdam University Medical Center, Amsterdam, Netherlands, 7Institute of Neurology, University College London, London, United Kingdom
    ASLDRO is digital reference object software for Arterial Spin Labelling. Here we present the development  and use of the DRO software, and its use in a sensitivity and uncertainty analysis of the single-subtraction equation for ASL perfusion quantification.
    a. Tissue segmentation volume from the input ground truth obtained from the ICBM 2009a Nonlinear Symmetric atlas12. Values in the table in b. are assigned to each of the regions for each quantity volume, relaxation times are for 3 Tesla. c. Example DRO output control, label and corresponding difference images. d. Four transversal MRI slices output from ExploreASL for two DROs with same perfusion but with and without motion. tSNR = temporal signal-to-noise ratio.
    Histograms (200 bins) of the randomly sampled input parameters (a-e). Labelling efficiency values were limited to a maximum of 1, resulting in an excess of samples with value 1.0. Histograms (200 bins) and fitted gaussians of the output mean CBF for grey matter (f) and white matter (g). Input-output graph showing the values of the mean CBF in each ROI for both the resampled ground truth, and the results form the Whitepaper equation calculation (h), error bars show one standard deviation.
  • Reproducibility and repeatability of quantitative pCASL measurements in a 3D-printed perfusion phantom
    Yiming Wang1, Limin Zhou1, Durga Udayakumar1,2, and Ananth J. Madhuranthakam1,2
    1Radiology, UT Southwestern Medical Center, Dallas, TX, United States, 2Advanced Imaging Research Center, UT Southwestern Medical Center, Dallas, TX, United States
    We assessed the reproducibility and repeatability of perfusion measurements using multi-PLD 2D pCASL over 5 weeks with a 3D-printed perfusion phantom. Intra-class correlation coefficients of measured perfusion and T1 are 0.96 and 0.94, indicating good reproducibility and repeatability.
    Figure 3. Signal intensities (open circle) and their regression analysis results (solid line) over 20 PLDs are shown for 3 runs of 2D pCASL in one session (a), and for all 5 sessions across 5 weeks (b). Curves were shifted horizontally slightly so that they all peak at LD+PLD = 5000 ms.
    Figure 2. perfusion difference images of 20 PLDs ranging from 0.2 s to 7.8 s from 3 runs (a, b and c) of 2D pCASL sequences in one session, compared against images from one run (d) of another session showing similar signal intensities.
Back to Top
Digital Poster Session - Arterial Spin Labelling: Applications
Diffusion/Perfusion
Wednesday, 19 May 2021 13:00 - 14:00
  • A quantitative multiparametric 18F-FPIA PET/MRI study for the characterization of primary brain gliomas
    Marianna Inglese1, Shah Islam1, Matthew Grech-Sollars1,2, Giulio Anichini3, James Davies4, Azeem Saleem4,5, Matthew Williams6,7, Kevin S O'Neill3, Adam D Waldman8, and Eric O Aboagye1
    1Surgery and Cancer, Imperial College London, London, United Kingdom, 2Imaging, Imperial College London Healthcare NHS Trust, London, United Kingdom, 3Imperial College London Healthcare NHS Trust, London, United Kingdom, 4Invicro Imperial College London, London, United Kingdom, 5Hull York Medical School, Faculty of Health Sciences, University of Hull, Hull, United Kingdom, 6Computational Oncology Group, Department of Surgery and Cancer, Imperial College London, London, United Kingdom, 7Institute for Global Health Innovation, Imperial College London, London, United Kingdom, 8Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
    A strong correlation was found between 18F-FPIA PET SUV and K1, and between the Ki and the MRI perfusion parameters rCBF and rCBV. By combining PET and MRI, the preliminary grade predictive vector provided 100% accuracy in tumour grade prediction.
    Figure 1: 18F-FPIA PET/MRI acquisition. Axial images of a representative patient of the standardised uptake value corrected by the tracer uptake in the whole blood (SUVc), the contrast agent plasma/interstitium transfer rate constant Ktrans and the extracellular extravascular volume fraction ve resulted from the post-processing of DCE-MRI data, the apparent diffusion coefficient (ADC) map from DWI-MRI acquisition, the relative contrast agent mean transit time (rMTT) derived from DSC-MRI and the cerebral blood flow (CBF) derived from the sequence of ASL.
    Figure 2: The least absolute shrinkage and selection operator (LASSO) was applied to extract a composite vector - the most significant tumour grade predictor - from 25 parameters determined by analysis of simultaneously acquired PET and MRI data. Five-fold cross-validation was performed to select lambda minimum to give the minimum cross-validated error (A). The weighted sum of 3 selected features (B) gave the Grade predictive Vector GpV, which showed an AUC and accuracy of 1 in the discrimination between low and high-grade gliomas (C).
  • Identification of IDH1 mutation status in glioblastoma using multi-delay 3D arterial spin labeling perfusion MRI: a pilot study
    Huilou Liang1,2, Lianwang Li3, Yuchao Liang3, Siqi Cai4, Jing An5, Yan Zhuo1,2,6, Lijuan Zhang4, Danny JJ Wang7, and Rong Xue1,2,8
    1State Key Laboratory of Brain and Cognitive Science, Beijing MRI Center for Brain Research, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China, 2University of Chinese Academy of Sciences, Beijing, China, 3Department of Neurosurgery, Beijing Tiantan Hospital of Capital Medical University, Beijing, China, 4Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China, 5Siemens Shenzhen Magnetic Resonance Ltd, Shenzhen, China, 6CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Beijing, China, 7Mark & Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States, 8Beijing Institute for Brain Disorders, Beijing, China
    In this study, we applied multi-delay 3D ASL with multi-parametric perfusion maps in GBM patients. Our results show that aCBV based relative perfusion parameters may provide a better performance in predicting IDH1 mutation status and is worthy of further verification in future studies.
    Figure 1. A 32-year-old woman (Patient #2) with IDH1-mutant GBM. ROIs of tumoral (red) and mirrored (green) regions were manually drawn on T2-FLAIR image referring to contrast-enhancing regions in post-contrast T1 image, and successively overlaid onto multi-parametric perfusion maps (lower row) to measure the maximum and mean values within ROIs. As indicated by red arrows, the CBF map shows obviously high values in regions of large vessels (confirmed by low ATT values) within tumor found in T2-FLAIR image, whereas the aCBV map doesn’t display elevated perfusion.
    Figure 2. A 57-year-old man (Patient #4) with IDH1-wild type GBM. ROIs of tumoral (red) and mirrored (green) regions were manually delineated on T2-FLAIR image and overlaid onto multi-parametric perfusion maps (lower row). Ring-enhancing lesions in post-contrast T1 image are coincident with ring-like regions with hyper-perfusion in CBF and aCBV maps. Prolonged ATT values especially in the occipital region are observed.
  • Intrasession reliability of arterial spin labeled MRI measured perfusion in GBM at 3T
    Limin Zhou1, Yiming Wang1, Marco Da Cunha Pinho1,2, Edward Pan3,4,5, Yin Xi1,6, Joseph A Maldjian1,2, and Ananth J Madhuranthakam1,2
    1Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX, United States, 2Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX, United States, 3Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, DALLAS, TX, United States, 4Department of Neurological Surgery, University of Texas Southwestern Medical Center, DALLAS, TX, United States, 5Harold C. Simmons Cancer Center, University of Texas Southwestern Medical Center, DALLAS, TX, United States, 6Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX, United States
    In this study, we compared the 3D TSE-CASPR measured perfusion with clinically available 3D GraSE in GBM patients at 3T. The results showed that 3D pCASL with TSE-CASPR is more robust to B0 inhomogeneities and has higher intrasession reliability than the clinical sequence, 3D pCASL with GraSE.
    Figure 1. Two runs of 3D pCASL with TSE-CASPR and three dynamics of 3D pCASL with GraSE in a 26-year old healthy volunteers (a) and a 71-year old GBM patient (b).
    Table 1. Intraclass correlation coefficient (ICC) and its 95% confidence interval (CI) for 2 runs of 3D pCASL with TSE-CASPR and 3 dynamics of 3D pCASL with GraSE.
  • The feasibility of 3D pcASL MRI with Multiple post-labeling delay times in evaluating subtypes of parotid gland tumors
    Lu Chen1, Guo-Yi Su1, Weiqiang Dou2, Yong Shen3, Fei-Yun Wu1, and Xiao-Quan Xu1
    1Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China, 2GE Healthcare, MR Research China, Beijing, P.R. China, Beijing, China, 3GE Healthcare, MR Enhanced Application China, Beijing, P.R. China, Beijing, China
    3D pcASL MRI, especially with short PLD was suggested to evaluate patients with parotid gland tumors in routine clinical practice.
    Figure 2. Representative images of PA (A-F), WT (J-L) and squamous cell carcinoma (M-R). The first column was the fat-suppressed T2-weighted image (A, J, M). The second (B, H, N), third (C, I, O), fourth (D, J, P), fifth (E, K, Q) and sixth (F, L, R) columns were TBF maps at different PLDs (PLD=1025ms, 1525ms, 2025ms, 2525ms, 3025ms), respectively. WT (H-L) showed relatively highest TBF, followed by squamous cell carcinoma (N-R) and PA (B-F). The unit of TBF is ml/100g/min. TBF= tumor blood flow; PLDs= post-labeling delay times; PAs= pleomorphic adenomas; WTs= warthin’s tumors.
    Figure 1. Line graphs showing the changes of TBF at different PLDs. TBF= tumor blood flow; PLDs= post-labeling delay times; PAs= pleomorphic adenomas; WTs= warthin’s tumors; MTs= malignant tumors.
  • Robust Implementation of a 3D Pulsed ASL Sequence for Assessment of Liver Perfusion
    Jörn Huber1, Daniel Hoinkiss1, and Matthias Günther1,2
    1Fraunhofer MEVIS, Bremen, Germany, 2University of Bremen, Bremen, Germany
    Arterial Spin Labeling (ASL) allows non-invasive assessment of liver perfusion. However, breathing motion, low perfusion rates and off-resonance are challenges in liver ASL. In this work, a robust implementation for a pulsed ASL sequence using 3D GRASE is given, addressing these challenges.   
    Figure 5: Perfusion weighted images using background suppression with two inversion pulses and the proposed multi-breathhold acquisition scheme and a free-breathing acquisition respectively. Arrows indicate areas with subtraction errors under free-breathing which are likely misinterpreted as perfusion signal.
    Figure 3: Three exemplary slices of perfusion-weighted images using two inversion pulses and different inflow times TI.
  • Depicting the developmental trajectories of brain cerebral blood flow using 3D ASL in children aged 28 days to 15 years.
    Peiyao Chen1, Chao Jin1, Xianjun Li1, Miaomiao Wang1, Congcong Liu1, Xiaoyu Wang1, Fan Wu1, Yuli Zhang1, Cong Tian1, Mengxuan Li1, Xiaocheng Wei2, and Jian Yang1
    1First Affiliated Hospital of Xi 'an Jiaotong University, Xi'an, Shaanxi, China, 2MR Research China, GE Healthcare, Beijing, China
    The estimated age with highest cerebral blood flow is earliest in the occipital lobe, followed by temporal and parietal lobe, at last in the frontal lobe. The perfusion of basal ganglia shows a U-shaped curve, which slowly increases with age.
    Table 1 Demographic data Note: Mean±SD
    Figure 1. Manual regions of interest(ROIs)were placed on the CBF map by using the aligned anatomical image as guidance. ROI was about 20-100mm2. (A)bilateral superior frontal gyrus and posterior central gyrus .(B)bilateral superior temporal gyrus.(C)bilateral occipital lobe(D)basal ganglia (bilateral thalamus, globus pallidus, putamen, caudate nucleus )
  • Brain perfusion in dementia with Parkinson's disease and Alzheimer’s disease: an arterial spin labeling MRI study
    Hongri Chen1, Weiqiang Dou2, and Wei yin Liu2
    1Dalian Medical University, Northern Jiangsu People’s Hospital, Yangzhou, China, Yangzhou, China, 2GE Healthcare, MR Research China, Beijing, P.R. China, Beijing, China
     The normalized cerebral blood flow (CBF) provided sensitive imaging-based markers that contribute to the differential diagnosis of the Parkinson's disease dementia (PDD) and Alzheimer’s disease (AD).
    Figure 1 Results of two‐sample t‐test between dementia patients and HC subjects. The above: PDD minus HC. The below: AD minus HC. Red color represents the increased perfusion, while the blue color represents the decreased perfusion (PFWE < 0.001).
    Figure 2 The normalized CBF differences between the PDD patients and the AD patients. Compared with AD, the PDD patients showed decreased CBF in the bilateral putamen and right SMA, as well as increased CBF in the right MTG, and right precuneus.
  • Relationship between global grey matter perfusion, damage and disability in multiple sclerosis
    Daniele Mascali1, Antonio Maria Chiarelli1, Ilona Lipp2,3, Anna Digiovanni4, Valentina Tomassini1,3,4, and Richard Geoffrey Wise1,3
    1Institute for Advanced Biomedical Technologies,Department of Neuroscience, Imaging and Clinical Sciences, "G. D'Annunzio University" of Chieti-Pescara, Chieti, Italy, 2Department of Neurophysics, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany, 3Cardiff University Brain Research Imaging Centre (CUBRIC) School of Psychology, Cardiff University, Cardiff, United Kingdom, 4MS Centre, Neurology Unit, SS. Annunziata University Hospital, Chieti, Italy
    We found that a reduced global grey matter hypo-perfusion in multiple sclerosis patients is associated with greater tendency to develop irreversible tissue damage and with worse clinical scores.
    Figure 3. Scatter plots of median GM CBF vs. clinical and cognitive scores. R and p-values are obtained via Pearson’s correlation.
    Figure 2. Scatter plots of median grey matter CBF vs. white matter lesion volumes, as assessed by T1-weighted (left panel) or T2-weighted (central panel) images. The right panel shows the correlation between CBF and the T1/T2 volume ratio. R and p-values are obtained via Pearson’s correlation.
  • ASL perfusion and disability in primary progressive MS: an observational cohort study
    Clara Delacour1, Ahmed-Ali El Ahmadi1, Gilles Brun1, Nadine Girard1,2, Christoph Heesen3,4, Arzu Ceylan Has3,4, and Jan-Patrick Stellmann2,3,4,5
    1Neuroradiology, APHM La Timone, Marseille, France, 2Aix-Marseille Univ, CNRS, CRMBM, UMR 7339, Marseille, France, 3Institute of Neuroimmunology and MS (INIMS), University Medical Centre Hamburg-Eppendorf, Hamburg-Eppendorf, Germany, 4Neurology, University medical centre Hamburg-Eppendorf, Hamburg-Eppendorf, Germany, 5APHM La Timone, CEMEREM, Marseille, France

    We notice no relevant change of brain perfusion over up to five years in primary progressive MS population cohort (77 patients). An association between higher regional perfusion rates and cognitive performance and hand functioning has been highlighted. 

    Figure 1
  • Reliability of Arterial Spin Labeling derived Cerebral Blood Flow measurements in Periventricular White Matter
    Sudipto Dolui1, Audrey P. Fan2,3, Moss Y. Zhao3, Greg Zaharchuk3, and John A. Detre1,4
    1Department of Radiology, University of Pennsylvania, Philadelphia, PA, United States, 2Departments of Biomedical Engineering and Neurology, University of California, Davis, CA, United States, 3Department of Radiology, Stanford University, Palo Alto, CA, United States, 4Department of Neurology, University of Pennsylvania, Philadelphia, PA, United States
    Cerebral blood flow in periventricular white matter measured using state-of-the-art arterial spin labeling showed high intra-session reproducibility and correlated with measurements from concurrently acquired [15O]-water PET, demonstrating it can be measured reliably.
    Figure 1: (A) Group averaged CBF obtained from 436 subjects (age=50.4±3.5 years, 54% female) from the NHLBI Coronary Artery Risk Development in Young Adults (CARDIA) study; (B) A periventricular white matter (PVWM) region of interest (ROI) obtained by thresholding the group averaged map in (A) to CBF<12.5ml/100g/min.
    Figure 2: (A)-(D): Scatter plots comparing the (A) whole brain (WB), (B) gray matter (GM), (C) white matter (WM) and (D) periventricular WM (PVWM) CBF obtained from the two sessions using single-PLD (shown in blue) and multi-PLD (shown in red) ASL. The black lines correspond to the unity line and the titles show the correlation coefficients. (E)-(H) and (I)-(L) show Bland-Altman Plots corresponding to (E & I) WB CBF, (F & J) GM CBF, (G & K) WM CBF and (H & L) PVWM CBF using the two sessions of single and multi-PLD data respectively. The wsCV values are in the titles of the plots.
  • Reduced Cerebral Blood Flow in Patients with Pulmonary Arterial Hypertension
    Bhaswati Roy1, Susana Vacas1, Kathy McCloy2, Rajan Saggar2, and Rajesh Kumar1,3,4,5
    1Anesthesiology, University of California Los Angeles, Los Angeles, CA, United States, 2Medicine, University of California Los Angeles, Los Angeles, CA, United States, 3Bioengineering, University of California Los Angeles, Los Angeles, CA, United States, 4Radiological Sciences, University of California Los Angeles, Los Angeles, CA, United States, 5Brain Research Institute, University of California Los Angeles, Los Angeles, CA, United States
    PAH patients show cognitive and mood deficits, and brain changes in those sites. However, the underlying cause of tissue damage in PAH patients remain unclear. We show reduced regional CBF in PAH patients over controls, and correlations between CBF and functional deficits in the condition.
    Figure 1: Brain regions with reduced CBF in PAH patients over control subjects. The sites with reduced CBF in PAH patients included the bilateral frontal white matter (a, b), left anterior (c), mid (d), and posterior (e), and right anterior (i) cingulate, left insula (f), bilateral corona radiata (g, h), and bilateral prefrontal cortices (j, k). All images are in neurological convention (L = left; R = right). Color bar indicates t-statistic values.
    Figure 2: Negative correlations emerged between CBF and mood symptoms and positive associations between cognition and CBF in PAH patients. Correlations appeared between mood scores and CBF values of the right insula (a, c), and left basal forebrain (b, d), and between MoCA scores and CBF values of the bilateral basal forebrain (e, f) and right insula (g). Figure conventions are same as in Figure 1.
  • Moving Towards Robust Quantification of Cerebrovascular Reactivity (CVR) using Pseudocontinuous Arterial Spin Labelling (pCASL)
    Colette C. Milbourn1, Thomas W. Okell2, and Nicholas P. Blockley1
    1The School of Life Sciences, University of Nottingham, Nottingham, United Kingdom, 2Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
    Cerebrovascular reactivity measured with Pseudocontinuous Arterial Spin Labelling (pCASL) systematically varies with different pCASL preparation parameters due to changes in blood velocity in the internal carotid and vertebral arteries.
    Figure 4 Comparison of CVR estimates. Box plots shows the median cerebrovascular reactivity (CVR) for each of the three pCASL (pseudocontinuous Arterial Spin Labelling) Parameter sets. CVR is the percentage change in cerebral blood flow per mmHg partial pressure of end-tidal carbon dioxide (PetCO2).

    Figure 3 ROI Selection and Blood Velocity Results. A: Phase contrast velocity region of interest (ROI) selection of left/right (L/R) internal carotid arteries (ICA) and vertebral arteries (VA). B: Average maximum velocity of feeding arteries (N=9, F=4, mean±s.d.) at three carbon dioxide (CO2) levels: hypocapnia (hypo, -10mmHg below baseline), normocapia (normo) and hypercapnia (hypo, +10mmHg above baseline).

  • Arterial Spin Labeling Can Identify Cerebrovascular Reactivity Deficit in Patients with Vasculopathy: A Pilot Study Using Simultaneous PET/MRI
    Moss Y Zhao1, Audrey P Fan2, David Chen3,4, Jia Guo5, Yosuke Ishii6, David Shin7, Mohammad Mehdi Khalighi1, Dawn Holley1, Kim Halbert1, Andrea Otte1, Brittney Williams1, Jun-Hyung Park1, Bin Shen1, Gary Steinberg8, and Greg Zaharchuk1
    1Radiology, Stanford University, Stanford, CA, United States, 2Biomedical Engineering and Neurology, University of California Davis, Davis, CA, United States, 3Medical Imaging, Taipei Medical University – Shuan-Ho Hospital, New Taipei City, Taiwan, 4Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, 5Bioengineering, University of California Riverside, Riverside, CA, United States, 6Neurosurgery, Tokyo Medical and Dental University, Tokyo, Japan, 7GE Healthcare, Melo Park, CA, United States, 8Neurosurgery, Stanford University, Stanford, CA, United States
    ASL is effective in measuring CVR and can become a diagnostic tool to predict the risk of stroke for vasculopathy patients.
    Figure 2: Structural and flow maps of an example patient. Occlusion are seen in right MCA and PCA territories in MRA of CoW and hyperintense focal spots in T2 FLAIR (marked by yellow arrows). In all PET and ASL modalities, CBF and CVR in the right MCA and PCA territories were lower than the right side.
    Figure 1: Structural and flow maps of an example patient. Severe stenosis and occlusion are seen in left ACA and MCA territories in MRA of CoW and hyperintense focal spots in T2 FLAIR (indicated by yellow arrows). In all PET and ASL modalities, CBF and CVR in the left ACA and MCA territories were lower than the right side.
  • A digital brain perfusion phantom for validation of ASL data post-processing software
    Chenyang Zhao1, Ze Wang2, and Danny Wang1
    1Laboratory of Functional MRI Technology (LOFT), Mark & Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, United States, 2Department of Diagnostic Radiology and Nuclear Medicine, School of Medicine, University of Maryland, Baltimore, MD, United States
    This work presented a digital brain perfusion phantom with flexible parameter settings to achieve various ASL configurations. It provides a reliable reference to validate ASL post-processing software.
    (a) A Sample slice for T1 weighted MPRAGE, Label, Control, and M0 image. (b) A sample slice for 5-PLD ASL images. The PLD ranges from 500 to 2500 ms with an interval of 500 ms. (c) The perfusion map of the checkerboard phantom at PLD = 1000 that is embedded in a lower slice of the phantom. (d) The simulated perfusion signal of GM and WM using a 5-delay 3D pCASL protocol.
    Resulted CBF (ml/100g/min) and ATT (ms) maps of the digital realistic phantom from 4 different ASL data processing software, including BASIL, Cereflow, MRI Cloud, and ASLtbx, under a normal SNR condition and a low SNR condition.
  • Association of Arterial Spin Labeling Global Metrics and Attentional Processes
    Shichun Chen1, Yakun Zhang1, Zongpai Zhang1, Wenna Duan1, George Weinschenk1, Brandon E. Gibb2, Wenming Luh3, and Weiying Dai1
    1Department of Computer Science, State University of New York at Binghamton, Binghamton, NY, United States, 2Department of Psychology, State University of New York at Binghamton, Binghamton, NY, United States, 3National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
    Brain global functional activity and global topology of brain networks, measured by dynamic arterial spin labeling, are associated with the P3 properties obtained from event-related potentials, indicating close correlation of brain global activity/connection efficiency and attention.
    Fig. 1. Relationship between the P3 parameters and numbers of correct response. (a) rank correlation of between P3 amplitude and numbers of correct response. (b) rank correlation between P3 amplitude and numbers of correct response
    Fig. 3. Relationship between P3 latency and global topology of brain networks. (a) significant rank correlation between P3 latency and global efficiency. (b) significant rank correlation between P3 latency and mean function connectivity. (c) significant rank correlation between P3 latency and characteristic path length.
  • Mapping water exchange rate change after caffeine uptake using 3D diffusion prepared arterial spin labeled perfusion MRI
    Qihao Zhang1, Jana Ivanidze2, Thanh Nguyen2, Pascal Spincemaille2, and Yi Wang1
    1Cornell University, New York, NY, United States, 2Weill Cornell Medical College, New York, NY, United States
    A 13% decrease in water exchange rate (kw) is observed after caffeine uptake, captured by a newly developed kw mapping sequence using arterial spin labeling (ASL) technique.
    (a) Bland Altman plot of the selected 10 brain regions of the 5 subjects. (b) The test-retest for kw of a subject.
    (a) and (c) CBF and kw maps before caffeine uptake. (b) and (d) CBF and kw maps after caffeine uptake. Averaged in whole brain, a 26% decrease in CBF and 13% decrease in kw are observed.
  • High spatio-temporal resolution 3D ASL renal perfusion with variable-density FSE and deep-learning reconstruction
    Manuel Taso1, Uri Wollner2, Arnaud Guidon3, Rafi Barda2, Christopher J Hardy4, Sangtae Ahn4, and David C Alsop1
    1Division of MRI research, Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States, 2GE Research, Herzliya, Israel, 3Global MR Applications and Workflow, GE Healthcare, Boston, MA, United States, 4GE Research, Niskayuna, NY, United States
    Arterial spin labeling (ASL) has proven to be a powerful research and clinical technique for functional imaging of tissues. This work explores the feasibility and performance of Deep-Learning based reconstruction for fast volumetric perfusion imaging with ASL.
    Figure 2 – comparison between L1-wavelet CS and DCI-net reconstructions at various rates
    Figure 1 – Illustration of the architecture of DCI-Net with 2D convolution.
  • Reproducibility of multiparametric MRI in transplanted kidneys
    Rebeca Echeverria-Chasco1,2, Marta Vidorreta3, Veronica Aramendia-Vidaurreta2,4, David Cano 1, Gorka Bastarrika2,4, Nuria Garcia-Fernandez2,5, Paloma L. Martin Moreno2,5, and Maria A. Fernandez-Seara2,4
    1Radiology, Clínica Universidad de Navarra, Pamplona, NE, Spain, 2IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain, 3Siemens Healthineers, Madrid, Spain, 4Radiology, Clínica Universidad de Navarra, Pamplona, Spain, 5Nephrology, Clínica Universidad de Navarra, Pamplona, Spain
    Reproducibility of multiparametric MRI (perfusion, diffusion and T1 mapping) was assessed in renal allograft
    Figure 1: Example of multiparametric MRI maps acquired during the two MRI exams: arterial spin labeling perfusion maps, diffusion coefficient (D) maps, pseudo-diffusion coefficient (D*) maps, perfusion fraction maps and longitudinal relaxation (T1) maps.
    Figure 2: a) Bland-Almant plots for the RBF (ml/min/100g), D (×10−3 mm2/s), D* (×10−3 mm2/s), PF (%) and T1 (ms) calculated in the cortex and in the medulla. b) Correlation between cortical mean values of renal blood flow and perfusion fraction averaged for the two exams, with a correlation coefficient r = 0.47.
  • Measurement of Pulmonary Perfusion under Expiratory and Inspiratory Breathing Conditions using PCASL-bSSFP Imaging at 1.5 Tesla
    Petros Martirosian1, Rolf Pohmann2, Martin Schwartz1,3, Thomas Kuestner4, Manuel Kolb4, Ahmed Othman4, Cecilia Zhang4, Klaus Scheffler2,5, Konstantin Nikolaou4, Fritz Schick1, and Ferdinand Seith4
    1Section on Experimental Radiology, University of Tübingen, Tübingen, Germany, 2Max Planck Institute for Biological Cybernetics, Tübingen, Germany, 3Institute of Signal Processing and System Theory, University of Stuttgart, Stuttgart, Germany, 4Department of Diagnostic and Interventional Radiology, University of Tübingen, Tübingen, Germany, 5Department of Biomedical Magnetic Resonance, University of Tübingen, Tübingen, Germany
    Pseudo-continuous arterial spin labeling (PCASL) is able to detect changes of parenchymal lung perfusion caused by alterations of the intrathoracic pressure. Perfusion signal measured under end-inspiratory condition was noticeably reduced as compared to end-expiratory breath-hold.
    Figure 2: PCASL perfusion-weighted images of three healthy volunteers acquired under expiratory, inspiratory and free-breathing conditions. One pair and twelve pairs of label-control images were measured in breath-hold and free-breathing examinations, respectively.
    Figure 1: Spatial arrangement of labeling and imaging planes on a sagittal image in PCASL measurements. The labeling plane (red) was positioned nearly perpendicular to the pulmonary trunk (yellow arrow) and images were acquired in coronal orientation (green). Time course of ECG-triggered PCASL sequence: labeling duration (τ) was limited to the systolic period and imaging was performed in diastole of successive cardiac cycle by adapting post-labeling delay (PLD).
  • Prostate Perfusion Mapping using Fourier-Transform based Velocity-Selective Pulse Trains:  Choice of Cutoff Velocity and Comparison with Brain
    Dapeng Liu1,2, Dan Zhu3, Wenbo Li1,2, and Qin Qin1,2
    1Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States, 2F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, United States, 3Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States
    Prostate blood flow and blood volume mapping using VSASL prepared by Fourier-transform based velocity-selective pulse trains were compared among different cutoff velocities (Vc). The results suggest that lower Vc of VSASL is demanded for prostate than for brain.
    Figure 2: Brain (a) and prostate (b) PWS and corresponding tSNR maps from one representative subject with four out of all ten slices. Both blood flow (CBF for brain and PBF for prostate) and blood volume (CBV for brain and PBV for prostate) maps were shown. M0 and DIR images were also shown above. Note that in the CBV PWS and tSNR images, the CSF in leteral ventricles display dark as it has negative signal due to a combination of long T2 values of CSF and the higher B1+ scale at the center of the brain.
    Figure 4: Averaged blood flow PWS (a) and its tSNR (b), blood volume PWS (c) and its tSNR (d), in ROIs of GM, WM and prostate from four subjects.