Diffusion in Cancer: Clinical Studies & Validation
Diffusion/Perfusion Thursday, 20 May 2021

Oral Session - Diffusion in Cancer: Clinical Studies & Validation
Diffusion/Perfusion
Thursday, 20 May 2021 12:00 - 14:00
  • Microstructural mapping with diffusion-time dependent diffusion MRI improves diagnosis of prostate cancer at 3T
    Dan Wu1, Kewen Jiang2, Yi-Cheng Hsu3, Yi Sun3, Yi Zhang1, and Yudong Zhang2
    1Biomedical Engineering, Zhejiang University, Hangzhou, China, 2Radiology, the First Affiliated Hospital with Nanjing Medical University, Nanjing, China, 3MR Collaboration, Siemens Healthcare Ltd., Shanghai, China
    In prostate cancer, fin and cellularity obtained from diffusion time-dependent diffusion MRI and IMPULSED model increased as Gleason score increased, while and diameter and Dex decreased. Cellularity achieved the highest diagnostic accuracy with an area-under-the curve of 0.96. 
    Figure 4: (A) Diagnostic performance of different microstructural markers to differeciate clinically significant (GS>1) and insignificant (GS≤1) cancers in terms of area-under-the curve (AUC), accuracy, sensitivity and specificity, based on a five-fold cross-validation using the linear discriminator. (B) Receiver-operating-curves (ROCs) of the different markers. (C) Classifying clinically significant (red) and insignificant (blue) cancers using the combined marker of the cellularity and DPGSE(30ms). The mis-classified cases are marked as “x”.
    Figure 1: Microstructural maps of the prostate tissues of Gleason score (GS) from 0-4. Intracellular fraction (fin), cell diameter (d), cellularity, and extracellular diffusivity (Dex) fitted from the IMPULSED model and the diffusivity maps from PGSE, OGSE (17Hz), and OGSE (33Hz) data were shown. The white contours and red arrows indicated the cancerous regions based on manual delineation.
  • Leveraging a multicompartmental signal model for improved classification of prostate-cancer bone metastases in whole-body DWI
    Christopher C Conlin1, Christine H Feng2, Leonardino A Digma2, Ana E Rodriguez-Soto1, Joshua M Kuperman1, Dominic Holland3, Rebecca Rakow-Penner1, Tyler M Seibert1,2,4, Anders M Dale1,3,5, and Michael E Hahn1
    1Department of Radiology, UC San Diego School of Medicine, La Jolla, CA, United States, 2Department of Radiation Medicine and Applied Sciences, UC San Diego School of Medicine, La Jolla, CA, United States, 3Department of Neurosciences, UC San Diego School of Medicine, La Jolla, CA, United States, 4Department of Bioengineering, UC San Diego Jacobs School of Engineering, La Jolla, CA, United States, 5Halıcıoğlu Data Science Institute, UC San Diego, La Jolla, CA, United States
    Multicompartmental modeling was applied to develop an empirical tissue classifier for identifying bone lesions in whole-body DWI. This classifier considerably outperformed one based on conventional ADC values.
    Figure 3: RSI cancer-likelihood map of a patient with prostate-cancer metastases in the pelvis and femur (cyan arrows), compared against conventional MR images. Bone lesions show a very high likelihood value [probability of being cancerous; P(cancer)] compared to surrounding normal tissue. Normal tissue is generally less pronounced on the likelihood map than on conventional MR images. False positive signal remains, however, in organs with dense cellular arrangement like the kidneys and brain.
    Figure 2: RSI signal distributions for normal tissue and bone lesions. The joint C1,C2 probability density functions (PDFs) are shown for normal control tissue (left) and bone lesions (middle). Both PDFs are shown after log transformation to better show less frequent combinations of C1 and C2. The posterior probability distribution on the right is derived from the PDFs and shows the likelihood of cancer [P(cancer)] given particular C1 and C2 values. High C1 signal in particular is indicative of cancer.
  • Biomimetic phantoms of impeded diffusion in prostate cancer using lipid nanoparticles
    Scott D. Swanson1, Thomas L. Chenevert1, Prasad R. Shankar1, Ted Lynch2, and Dariya I. Malyarenko1
    1Department of Radiology, University of Michigan, Ann Arbor, MI, United States, 2CIRS, Norfolk, VA, United States
    A biomimetic phantom has been developed with a range of multi-compartment diffusion parameters similar to those observed in cancerous and health prostate tissue.
    Figure 1. Schematic of biomimetic lipid nanoparticle phantom to reflect multi-compartment contribution to DWI voxel signal in PCa (GS7 histology) and example fit parametric maps for diffusion-kurtosis model. Sizes of the lipid nanoparticles can be controlled to be between 200 and 2,000 nm, depending on the composition of the nanoparticle shell.
    Figure 3. Scatter plot of apparent kurtosis, Ka, versus apparent diffusion, Da, for in vivio PCa and selected diffusion-kurtosis phantom materials. The range of values found in PCa GS6 and GS7 lesions for Da and Ka (estimated by fitting DWI decay curves to the isotropic diffusion kurtosis model) can be spanned by fabrication of lipid nano-materials with appropriate physical properties.
  • Evaluate the micro-vascular invasion in HCC with a Fractional Order Calculus DWI Model
    Xiuzhong Yao1, Yunfei Zhang2, Mengsu Zeng1, and Yongming Dai2
    1Zhongshan Hospital affiliated to Fudan University, Shanghai, China, 2Central Research Institute, United Imaging Healthcare, Shanghai, China
    FROC-DWI holds great potential in noninvasively and accurately evaluating the MVI in patients with HCC.
    Figure 2. Representative FROC-derived parametric images of one patient with micro-vascular invasion.
    Fig. 4: Diagnostic performance of FROC-derived parameters for predicting the MVI in patients with HCC.
  • DR-HIGADOS: a new diffusion-relaxation framework for clinically feasible microstructural imaging of the liver
    Francesco Grussu1, Ignasi Barba2, Kinga Bernatowicz1, Irene Casanova-Salas3, Alba Escriche Villarroya4, Natalia Castro3, Emanuela Greco4, Juan Francisco Corral5,6, Marta Vidorreta7, Manuel Escobar Amores5,6, Núria Roson5,6, Xavier Merino5,6, Richard Mast5,6, Nahúm Calvo‐Malvar5,8, Joaquin Mateo3, Paolo Nuciforo9, María Abad4, Josep R. Garcia-Bennett8, and Raquel Perez-Lopez1,6
    1Radiomics Group, Vall d'Hebron Institute of Oncology, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain, 2NMR Lab, Vall d'Hebron Institute of Oncology, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain, 3Prostate Cancer Translational Research Group, Vall d'Hebron Institute of Oncology, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain, 4Cellular Plasticity and Cancer Group, Vall d'Hebron Institute of Oncology, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain, 5IDI (Institut de Diagnòstic per la Imatge), Catalonia, Spain, 6Department of Radiology, Hospital Universitari Vall d'Hebron, Barcelona, Spain, 7Siemens Healthineers, Madrid, Spain, 8Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, Spain, 9Molecular Oncology Group, Vall d'Hebron Institute of Oncology, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
    We introduce DR-HIGADOS, a new liver diffusion-relaxation MRI technique providing indices of cell size and cellularity. DR-HIGADOS is shown to be feasible on multi-vendor clinical scans, and preliminary MRI-histology correlations confirm its biological specificity.
    Figure 1: overview of the two-step DR-HIGADOS framework. In the first step, an extension of the IVIM model is fitted to diffusion-relaxation measurements. In the second step, model parameters are mapped to biophysical properties (e.g. average intrinsic cell diffusivity and size; cellularity), using information derived from Monte Carlo simulations.
    Figure 2: parametric maps provided by DR-HIGADOS in vivo on two MRI vendors. Left to right: non-DW image, vascular signal fraction f , vascular diffusivity and T2 (Dv and T2V), tissue diffusivity, kurtosis excess and T2 (DT, KT and T2T), average cell diffusivity and size (D0 and L), cellularity C. Top to bottom: healthy volunteer on Ingenia system; healthy volunteer on Avanto system; patient on Avanto system.
  • Ex vivo and In vivo Diffusion Weighted MRI highlights the Microarchitecture of mouse Pancreatic Intraepithelial Neoplasia
    Carlos Bilreiro1,2, Francisca F Fernandes1, Rui V Simões1, Mireia Castillo-Martin1,3, Andrada Ianus1, Cristina Chavarrias1, Celso Matos1,2, and Noam Shemesh1
    1Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, Portugal, 2Department of Radiology, Champalimaud Clinical Centre, Lisbon, Portugal, 3Department of Pathology, Champalimaud Clinical Centre, Lisbon, Portugal
    We developed a diffusion-MRI (dMRI) approach for mapping pancreatic cancer precursor lesions in transgenic mice via ex vivo dMRI Microscopy with histological validation, and applied the methods in vivo. Highly sensitive contrasts were discovered.
    MR Microscopy, Pdx1-Cre;KrasG12D mouse pancreas. A – PanIN in ADM background. B – Area of PDAC. As observed in the previous example, PanIN (yellow arrows) are most conspicuous with high b values and longer echoes, with high peripheral anisotropy values. PDAC (green arrows) presents with similar contrasts, but with lower anisotropy values in its solid central portions, possibly due to a different cellular and fibrotic content.
    In vivo dMRI. (A) The healthy pancreas has low signal intensity in DWI and low anisotropy values. (B) A large abdominal mass is seen, compatible with PDAC (green arrows), with high DWI signal intensity and high anisotropy values in its solid areas. (C) Much smaller lesions, compatible with PanIN (yellow arrows), are depicted with similar contrasts. (D) Similar lesions are observed (yellow arrows), along with diffuse pancreatic changes compatible with PanIN/ADM (blue arrows).
  • Looking Inside a Voxel through the Lenses of Non-Gaussian Diffusion MRI: Correlation between Imaging- and Histology-based Tissue Heterogeneity
    Muge Karaman1,2, Guangyu Dan1,2, Lingdao Sha3, Tingqi Shi1, Weiguo Li4,5, Dan Schonfeld2,3,6, Tibor Valyi-Nagy7, and X. Joe Zhou1,2,8
    1Center for Magnetic Resonance Research, University of Illinois at Chicago, Chicago, IL, United States, 2Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, United States, 3Department of Electrical and Computer Engineering, University of Illinois at Chicago, Chicago, IL, United States, 4Research Resources Center, University of Illinois at Chicago, Chicago, IL, United States, 5Department of Radiology, Northwestern University, Chicago, IL, United States, 6Department of Computer Science, University of Illinois at Chicago, Chicago, IL, United States, 7Department of Pathology, University of Illinois at Chicago, Chicago, IL, United States, 8Departments of Radiology and Neurosurgery, University of Illinois at Chicago, Chicago, IL, United States
    The machine-learning classifier that quantifies histology-based heterogeneity achieved an accuracy of 90% and specificity of 94%. The CTRW parameters, Dm, α, β showed significant differences (p<0.05) among heterogeneity levels in normal and glioma tissue except β in the glioma specimen.
    Figure 1: Workflow of the study.
    Figure 3: T1 image in a), CTRW maps, Dm (in mm2/msec) in b), α in c), β in d), and co-registered histology-based blended probability maps in e) for three representative slices from the NAs. The blended probability maps in e) were generated by determining the “assigned” heterogeneity level (AHL) as the one with the highest probability; and displaying AHL’s probability with a customized color bar. For example, a yellow pixel with a value of 0.90 was predicted to have a heterogeneity level of L3 with a 90% probability. The L2 and L3 ROIs given in f) were drawn with the guidance of AHLs.
  • Diffusion MRI study of chemoradiation treatment response in patients with HPV positive oropharyngeal carcinoma
    Sungheon Gene Kim1, Mehran Baboli1, Justin Fogarty2, Steven H. Baete2, Joseph Kim3, Paulina Galavis3, Moses Tam3, Kenneth Hu3, and Elcin Zan2
    1Radiology, Weill Cornell Medical College, New York, NY, United States, 2Radiology, New York University School of Medicine, New York, NY, United States, 3Radiation Oncology, New York University School of Medicine, New York, NY, United States
    Diffusion MRI at long diffusion times (200-700 ms) found that the patients with less than 40% nodal volume shrinkage had significantly higher diffusivity at pretreatment and lower kurtosis at week4 than the other patients with better response.
    Figure 1: Example b=0 images of a non-deescalated and deescalated patients at pre-treatment and week 4 into the treatment. The metastatic lymph nodes are noted by arrows.
    Figure 3: Box-whisker plots of diffusivity and diffusion kurtosis of metastatic lymph nodes measured at long diffusion times. Black square boxes are for non-deescalated patients (n=6) and red boxes with notches are for deescalated patients (n=12).
  • Relating tumor site-specific volume and ADC changes following neoadjuvant chemotherapy to histopathology in epithelial ovarian cancer
    Jessica M Winfield1,2, Jennifer C Wakefield1,2, James D Brenton3,4,5, Khalid AbdulJabbar6,7, Antonella Savio8, Susan Freeman9, Erika Pace1,2, Kerryn Lutchman-Singh10, Katherine M Vroobel8, Yinyin Yuan6,7, Susana Banerjee11, Nuria Porta12, Shan E Ahmed Raza6,7,13, and Nandita M deSouza1,2
    1MRI Unit, Royal Marsden NHS Foundation Trust, London, United Kingdom, 2Division of Radiotherapy and Imaging, Institute of Cancer Research, London, United Kingdom, 3Cancer Research UK Cambridge Institute, Cambridge, United Kingdom, 4Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom, 5Department of Oncology, University of Cambridge, Cambridge, United Kingdom, 6Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom, 7Division of Molecular Pathology, The Institute of Cancer Research, London, United Kingdom, 8Department of Pathology, Royal Marsden NHS Foundation Trust, London, United Kingdom, 9Department of Radiology, Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom, 10Swansea Gynaecological Oncology Centre, Swansea Bay University Health Board, Singleton Hospital, Swansea, United Kingdom, 11Gynaecology Unit, Royal Marsden NHS Foundation Trust, Sutton, United Kingdom, 12Clinical Trials and Statistics Unit, The Institute of Cancer Research, London, United Kingdom, 13Department of Computer Science, University of Warwick, Warwick, United Kingdom
    Repeatability of ADC estimates in primary and metastatic tumor sites in epithelial ovarian cancer, and correlation with histopathological metrics (residual tumor and necrosis) after neoadjuvant chemotherapy.
    Figure 5: Scatter plots of post-NAC (pre-operative) ADCmedian vs. tumor cell fraction (left panel) and change in ADCmedian after three or four cycles of neoadjuvant chemotherapy and percentage necrosis.
    Figure 4: Normalised probability density functions for ADC estimates from ovarian, omental, and peritoneal lesions, and lymph nodes at baseline (pre-NAC) and after three or four cycles of neoadjuvant chemotherapy (post-NAC, pre-operative).
  • Endometrial Carcinoma: Assessment of Histological Features Based on Amide Proton Transfer-weighted Imaging and Diffusion Kurtosis Imaging
    Nan Meng1, Zhun Huang2, Ting Fang1, Pengyang Feng2, Xuejia Wang3, Dongming Han3, Kaiyu Wang4, and Meiyun Wang*1
    1Department of Radiology, Zhengzhou University People’s Hospital & Henan Provincial People’s Hospital, Zhengzhou, China, 2Department of Radiology, Henan University People’s Hospital & Henan Provincial People’s Hospital, Zhengzhou, China, 3Department of MRI, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China, 4GE Healthcare, MR Research China, BeiJing, China
    Our results showed that both Amide proton transfer-weighted imaging (APTWI) and Diffusion kurtosis imaging (DKI) were effective in the assessment of endometrial carcinoma in terms of clinical type, histological grade, subtype, and Ki-67 index.
    Figure.1. Images in a 49-year-old woman with type I, low-grade (grade 1) EA (arrowheads, Ki-67 = 30%). (a) DWI original maps (b = 1000 s/mm2), (b) Pseudo colored maps of Kapp, (c) Pseudo colored maps of Dapp, (d) APTWI original maps, (e) Pseudo colored maps of MTRasym (3.5ppm), (f) Pathological images (original magnification, ×100).
    Figure.1. Images in a 49-year-old woman with type I, low-grade (grade 1) EA (arrowheads, Ki-67 = 30%). (a) DWI original maps (b = 1000 s/mm2), (b) Pseudo colored maps of Kapp, (c) Pseudo colored maps of Dapp, (d) APTWI original maps, (e) Pseudo colored maps of MTRasym (3.5ppm), (f) Pathological images (original magnification, ×100).
Back to Top
Digital Poster Session - Microstructure: Models, Sampling & Analysis
Diffusion/Perfusion
Thursday, 20 May 2021 13:00 - 14:00
  • Recovering almost everything that diffusion could reveal
    Evren Özarslan1,2
    1Department of Biomedical Engineering, Linköping University, Linköping, Sweden, 2Center for Image Science and Visualization, Linköping University, Linköping, Sweden
    A new diffusion-encoding and data analysis framework is introduced with which the true diffusion propagator can be measured. The technique is sensitive to the structural parameters of the pore space.
    Fig. 3 Simulations for two compartments separated by a membrane of permeability $$${w}=0.6\,\mu m/ms$$$. Compartment sizes: $$$L_L=4.5\,\mu m$$$ (left), $$$L_R=5.5\,\mu m$$$ (right), diffusivities: $$$D_L=3\,\mu m^2/ms$$$, $$$D_R=2\,\mu m^2/ms$$$. Time-scales: $$$\tau_L=L_L^2/\pi^2D_L$$$, $$$\tau_R=L_R^2/\pi^2D_R$$$, $$$\tau_{ex}=\sqrt{D_L D_R}/{w}^2$$$. Top to bottom: true propagator, estimated propagator, EAP. Left three columns: near-ideal parameters. Last column: $$$\delta_1=200\,ms,\delta_{2,3}=1.2\,ms$$$, and $$$G_{max}=10\, T/m$$$.
    Fig. 1 (a) Effective gradient waveform of the Stejskal-Tanner sequence [1]. The signal can be transformed into the ensemble average propagator [2]. (b) Effective gradient waveform of the sequence by Laun et al [3], which can be utilized to obtain the long-time form of the diffusion propagator. (c) Effective waveform for one realization of the experiment considered. Here, $$$-\mathbf q-\mathbf q’$$$, $$$\mathbf q$$$, and $$$\mathbf q’$$$ are the signed areas under the first, second, and third gradient pulses, respectively. The signal can be transformed into the actual propagator.
  • Validation of between-bundle differences and within-bundle continuity of microstructural indices in ex vivo human brain tissue
    Robert Jones1, Chiara Maffei1, Qiuyun Fan1, Jean Augustinack1, Barbara Wichtmann2, Aapo Nummenmaa1, Susie Huang1, and Anastasia Yendiki1
    1Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States, 2Department of Radiology, University Hospital Bonn, Bonn, Germany, Bonn, Germany
    Ex vivo diffusion MRI signals exhibit bi-exponential decay as a function of q-value for both motor and association fibers. Parallel and perpendicular diffusion coefficients vary smoothly along each bundle, with marked differences between bundles.
    Figure 2. Decay curves for the diffusion signals parallel (light shade) and perpendicular (dark shade) to the fiber axis plotted as a function of q-value, for ROIs in the SLF (green) and CST (blue). Curves are shown for diffusion times of 30 ms (solid lines, filled markers) and 50 ms (dashed lines, open markers).
    Figure 1. Left: Sample extracted from a coronal slab of a human hemisphere. Right: Locations of SLF (left) and CST (right) ROIs from one representative coronal slice, overlaid on the primary fiber orientation vectors and corresponding FA map from GQI reconstruction.
  • Oscillating Gradient Spin Echo-Based Time-dependent Diffusivity Reflects Regional Microstructure Differences in Human White Matter
    Ante Zhu1, J. Kevin DeMarco2,3, Robert Y. Shih2,3, Radhika Madhavan1, Tim Sprenger4, Chitresh Bhushan1, Maureen Hood2,3, Luca Marinelli1, Vincent B. Ho2,3, and Thomas K.F. Foo1
    1GE Global Research, Niskayuna, NY, United States, 2Uniformed Services University of the Health Sciences, Bethesda, MD, United States, 3Walter Reed National Military Medical Center, Bethesda, MD, United States, 4GE Healthcare, Stockholm, Sweden
    The axonal fiber parallel diffusivity (PD) measured by oscillating gradient spin echo is left-right symmetric in healthy corpus callosum. The genu and splenium showed regional difference of time-dependent PD, indicating different tissue tortuosity and randomly distributed restrictions.
    Figure 2. Segmented parcels #1-#13 of the corpus callosum; and parallel diffusivity measurements at different OGSE frequencies of the corpus callosum. Different colors represent different subjects. L: left; R: right.
    Figure 1. OGSE with different frequencies and PGSE with flattened waveforms used for diffusion imaging (top), and the corresponding diffusion spectrum (bottom).
  • A tale of two frequencies: optimizing oscillating gradients for frequency dependent differential kurtosis mapping
    Kevin B Borsos1,2, Desmond HY Tse2, Paul I Dubovan1,2, and Corey A Baron1,2,3
    1Department of Medical Biophysics, Western University, London, ON, Canada, 2Centre for Functional and Metabolic Mapping, Western University, London, ON, Canada, 3Robarts Research Institute, Western University, London, ON, Canada
    We present an optimized oscillating gradient protocol to observe the difference in apparent kurtosis between OGSE and PGSE acquisitions in vivo without the requirement of a high-performance gradient insert.
    Figure 4: Differential ADC (A, C) and differential kurtosis (B, D) maps for both subjects produced from the images shown in Figure 3. ΔD and ΔK maps are generated from the subtraction of PGSE images from OGSE images using ADC maps and apparent kurtosis maps respectively. The distortions in the frontal lobe of Subject 1 are due to B0 inhomogeneity.
    Figure 1: Gradient waveforms (A, C) and spectral densities (B, D) of a conventional N = 2 cosine OGSE waveform (A) and our abbreviated oscillating gradient waveform (C). Here the second diffusion gradient is inverted to reflect the effect of the refocusing RF pulse (not shown). A reduction of the total diffusion gradient duration is apparent for the new waveform (C) compared to OGSE (A) at the same oscillation frequency of 25 Hz. Comparison of the respective spectra in (B) and (D) shows similar spectral selectivity and demonstrates zero DC spectral component for both waveforms.
  • When Averaging Only Gets You So Far: Repulsion and Peanut Squashing in Diffusion Tensor MRI
    Samuel Bryce Jones1, Emre Kopanoglu2, Chantal Tax2, and Derek Jones2
    1Radyr Comprehensive School, Cardiff, United Kingdom, 2CUBRIC, School of Psychology, Cardiff, United Kingdom
    We explore the extent to which reductions in SNR can be accounted for by signal averaging in diffusion tensor MRI experiments. Different metrics have different tolerances to reductions in SNR. We discuss how the rectified noise-floor and eigenvalue repulsion underpin these different results.
    FIGURE 3 - The 'classical' view of impact of SNR on eigenvalues, mean diffusivity, radial diffusivity and fractional anisotropy. In this simulation, the number of averages was not increased to compensate for the loss of SNR. This result provides useful insights into the impact of noise. E.g. we see both over-estimation of the largest eigenvalue (when FAtrue is low) and under-estimation (FAtrue is high), in line with previous literature1,2 on repulsion and squashing peanuts
    FIGURE 2 - Results of pair-wise t-tests, between all possible [SNR, N] couples. Blue entries show where the hypothesis that the samples come from the same distribution cannot be rejected at the p < 0.05 level. Yellow entries are where we have to reject this hypothesis and deem that the means are significantly different. Note the pattern depends both on FAtrue and on the metric itself.
  • Computing the Orientational-Average of Diffusion-Weighted MRI Signals: A Comparison of Different Techniques
    Maryam Afzali1, Hans Knutsson2,3, Evren Özarslan2,3,4, and Derek K Jones1,5
    1Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Cardiff, United Kingdom, 2Department of Biomedical Engineering, Linköping University, Linköping, Sweden, 3Center for Medical Image Science and Visualization, Linköping University, Linköping, Sweden, 4These authors share last authorship, Linköping, Sweden, 5These authors share last authorship, Cardiff, United Kingdom
    Orientationally-averaged diffusion-weighted signal can be computed using arithmetic averaging, weighted signal averaging; spherical harmonic; and Mean Apparent Propagator MRI (MAP-MRI) are compared. At low SNR and low number of data points MAP based approaches are better than the others.

    Fig. 1. The results from 488 samples for both shelled (61×8) and non-shelled point sets 18 in the presence of Gaussian noise. (a) the mean and std of the estimated signal versus b-value using MAP-MRI method with Nmax = 6 for five different noise floors, σg, and three different dispersion values, κ. The thickness of the blue band is twice the standard deviation of the signal estimates and its center is the mean. The dashed black line shows the ground truth and the red dots and bars show the results of the SH (L = 6). (b) the mean and std of the d1 and d2 measures for different methods.

    Fig. 5. (a) the estimated d1 and d2 for three different κ values, 344 (43×8) point sets in the presence of five different Gaussian noise levels. (b) the results of MAP-based interpolation of the orientationally-averaged data from Knutsson method (`MAP, Knutsson, s8' and `MAP, Knutsson, s43') on 43×8 shelled Lebedev 25 and the interpolation of original data (before averaging) on shelled (`MAP, s8' and `MAP, s43') and non-shelled point sets (`MAP, ns8' and `MAP, ns43') 18.
  • Towards a computational framework for task-driven experimental design
    Sean C Epstein1, Timothy J.P. Bray2, Margaret A. Hall-Craggs2, and Hui Zhang1
    1Department of Computer Science & Centre for Medical Image Computing, University College London, London, United Kingdom, 2Centre for Medical Imaging, University College London, London, United Kingdom
    A method enabling task-specific assessment of DWI experimental design choices. Validation with clinical data confirms its accuracy; illustrative use cases demonstrate its advantages of current computational experimental design practice.
    Figure 1 – Graphical overview of proposed CED assessment pipeline
    Figure 2 – Simulated (top row) vs. clinical (middle row) ROC curves for Zhao’s dataset7, for Tasks 1-3. The bottom row compares clinical AUC values to the distribution of simulated AUC values obtained by sub-sampling the pipeline dataset to match Zhao’s sample sizes. All ROC curves are qualitatively similar; the relative performance (AUC values) of different IVIM parameters are equal; all AUC values are in numerical agreement once clinical sample size is considered
  • Multi-component diffusion technique acquisition protocol optimization for different microstructure models
    Tommaso Ciceri1, Alberto De Luca2,3, Filippo Arrigoni1,4, and Denis Peruzzo1
    1NeuroImaging Unit, Scientific Institute IRCCS Eugenio Medea, Bosisio Parini, Italy, 2Neurology Department, UMC Utrecht, Utrecht, Netherlands, 3PROVIDI Lab, Image Sciences Institute UMC Utrecht, Utrecht, Netherlands, 4Radiology Unit, Fatebenefratelli Hospital, Milan, Italy
    A minimum, generic diffusion MRI acquisition scheme supporting the reliable application of multiple common estimation methods is investigated.
    Figure 2: Example of the parametric maps obtained using the whole available dataset (REFERENCE) and the minimum acquisition scheme (MAS), and the percentage error maps (ERR [%]) for the main parameters for each model.
    Figure 1: Frequency of appearance for each shell in the minimum acquisition schemes computed in the population. Red shells represent the population minimum acquisition scheme (MAS) for each model.
  • Optimizing DWI b-value Sampling for Accurate Metabolic and Cytometric Parameter Extraction: Activity MRI [aMRI]
    Xin Li1, Eric M. Baker1, Brendan Moloney1, Cory Wyatt1, Eric Baetscher1, Erin W. Gilbert2, Charles S. Springer1, Alexander R. Guimaraes1,3, and William D. Rooney1
    1Advanced Imaging Research Center, Oregon Health & Science University, Portland, OR, United States, 2Surgery, Oregon Health & Science University, Portland, OR, United States, 3Diagnostic Radiology, Oregon Health & Science University, Portland, OR, United States
    Using seven b-value acquisitions as an example, the optimal maximum DWI b-value for pancreas tail tissue is found to be generally under 3,000 s/mm2.  Evenly-spaced b-value strategies are often sufficient.  
    Figure 3. Contour plots of ln (1- a) for different bmax (from 2,000 to 7,000) and different b-spacing power (r) values for four different SNRs (single real or imaginary channel) of 50 (A), 100 (B), 200 (C), 300 (D). The log spacing option (gray arrows) was appended to the far right to the r values in each panel with white shading for differentiation. For normal pancreatic tail tissue, a bmax under 3000 s/mm2 is sufficient for practical achievable SNR and even b-spacing pattern is often among the optimal choices. See Fig. 4 for more details.
    Figure 4. The numerically determined radius (R) of curvature reciprocal for the “true” DWI curve (Fig. 1 solid curve) is plotted against the b-values in panel A. When 1/R approaches zero, the b-space decay approaches a straight line. Panel B plots the derivative of (1/R) with respect to b. It quantifies how quickly the curvature in panel A changes with respective to b. For this case, the most dramatic change occurs when b < 3,000 s/mm2, matching the Fig. 3 simulation results. (The ordinate units are not given.)
  • Data driven algorithm for multicomponent T2 analysis based on identification of spatially global sub-voxel features
    Noam Omer1, Neta Stern1, Tamar Blumenfeld-Katzir1, Meirav Galun2, and Noam Ben-Eliezer1,3,4,5,6
    1Department of Biomedical Engineering, Tel-Aviv University, Tel-Aviv, Israel, 2Department of Computer Science and Applied Mathematics, Weitzman institute of science, Rehovot, Israel, 3Department of Orthopedics, Shamir Medical Center, Zerifin, Israel, 4Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel, 5Sagol School of Neuroscience, Tel-Aviv University, Tel-Aviv, Israel, 6Center for Advanced Imaging Innovation and Research (CAI2R), New-York University Langone Medical Center, New York, NY, United States
    A novel data-driven approach for multicomponent analysis is introduced. This technique harnesses the statistical power of identifying global anatomical features prior to analyzing each voxel locally, offering reproducible estimation of myelin content in vivo.
    Figure 2. Repeatability test of the new mcT2 algorithm on in vivo brain data. Parametric maps of white matter (WM) segments from 3 consecutive scans of the same subject using the suggested algorithm. (a-d) mask and myelin water fraction (MWF) maps of genu of corpus callosum (GCC). (e-h) mask and MWF maps of splenium of corpus callosum (GCC). (i-k) mask and MWF maps of the cortical WM segment (CSEG). MWF maps are presented with the same color scale and on top of a T2 map presented in gray scale.
    Figure 1. Flowchart describing how to identify spatially global microscopic features for a given white matter (WM) segment. (a) A correlation-based probability score is computed between WM segment data and dictionary elements. (b-c) All scores are normalized and powered by β to prioritize the signals according to their probability to be found within the segment. (d) Summation of all signals scores to assign a global score for each dictionary element. (e) Selection of a L elements with the highest score.
  • Ultra-strong gradient diffusion MRI at 7T with a head insert
    Chantal Tax1,2, Edwin Versteeg1, Dennis J.W. Klomp1, Martijn F. Froeling1, Alberto de Luca1, and Jeroen C.W. Siero1,3
    1University Medical Center Utrecht, Utrecht, Netherlands, 2CUBRIC, Cardiff University, Cardiff, United Kingdom, 3Spinoza Centre for Neuroimaging Amsterdam, Amsterdam, Netherlands
    This work focuses on the importance of strong gradients - here provided by a gradient head insert - for high SNR and short TE diffusion imaging at 7T. Proof-of-principle images show that a short TE (21 ms) at a b-value of 1000 s/mm2 is achievable using an EPI-readout. 
    Maximum achievable b-value as a function of TE, for the gradient head insert (solid lines) and body gradients (dashed lines). RF pulse durations of 4 and 6 ms were assumed. The curves are shown for varying readout times [0, 5, 10, 15] ms e.g. depending on resolution and FOV, with a readout time of 0 ms representing a readout strategy starting at the centre of k-space (e.g. spiral imaging5,14).
    a) Signal decay as a function of TE at 3T (blue) and 7T (red), with tTE the threshold where the signals are equal. A linear increase in signal was assumed as a function of field strength and the apparent T2 was set to 50 and 77 ms at 7T and 3T respectively. b) tTE as a function of other settings for the relative signal gain at 7T - which can depend on e.g. body noise11 - and T2.
  • Brain microstructure at 1.5mm resolution via RMT reconstruction on a high-slew rate MAGNUS system
    Gregory Lemberskiy1, Santiago Coelho1, Thomas K.F. Foo2, Radhika Madhavan2, Luca Marinelli2, Jaemin Shin3, Els Fieremans1, and Dmitry S Novikov1
    1Radiology, NYU School of Medicine, New York, NY, United States, 2GE Research, Niskayuna, NY, United States, 3GE Healthcare, New York, NY, United States
    We present a near distortion-free and noise-free multishell diffusion neuro protocol at 1.5 mm enabled by the high performance MAGNUS head gradient coil reconstructed with random matrix theory denoising at the coil level. We showcase precise diffusion, kurtosis, and standard model maps. 
    Standard Model. We show parametric maps for Standard Model parameters: axonal volume fraction $$$f$$$, free water fraction $$$f_w$$$, angular dispersion $$$\theta_{disp} = \cos^{-1}\sqrt{(2p_2+1)/3}$$$, axonal (neurite) diffusivity $$$D_a$$$, longitudinal extra-axonal diffusivity $$$D_e^\parallel$$$, and radial extra-axonal diffusivity $$$D_e^{\perp}$$$. Streamline tractograms are are colored by their correspsonding SM parameters.
    RMT Reconstruction. (A) 5/32 coils and their corresponding residuals are shown before and after noise removal via RMT. Properties of the removed normalized residuals $$$r$$$ are evaluated via (B,C) histograms $$$p(r)$$$, and (D,E) power spectrum analysis. (F) Standard and RMT reconstructions following parallel imaging and coil combination of a $$$b=4000\,$$$s/mm$$$^2$$$ image are displayed for several slices.
  • Comparison of DCE-MRI and FEXI in the measurement of vascular water exchange in high-grade glioma
    Zejun Wang1, Bao Wang2, Yingchao Liu3, and Ruiliang Bai1,4
    1Key Laboratory of Biomedical Engineering of Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China, 2Department of Radiology, Qilu Hospital of Shandong University, Jinan, China, 3Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China, 4Department of Physical Medicine and Rehabilitation, Interdisciplinary Institute of Neuroscience and Technology, The Affiliated Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
    We compared shutter speed (SS) DCE-MRI and filter-exchange imaging (FEXI) for vascular water exchange measurement in high-grade glioma. Our results demonstrated consistent vascular water exchange assessments by SS DCE-MRI and FEXI in both normal-appearing white matter and tumor.
    Figure 4: The kbo map from DCE-MRI and AXR map from FEXI on the same slice. kbo’s underlay is enhanced image, and AXR’s underlay is T2-weighted image. The kbo and AXR shows similar spatial patterns as pointed with red arrows.
    Figure 2: (a) The illustration of FEXI sequence. (b) At equilibrium, MR-visible water molecules (white dots) are located in blood with fast diffusivity and interstitium with show diffusivity. Signal from fast diffusing intravascular water is suppressed after filter applied, indicated by black dots, leading to a reduction in the ADC′. After tm, water molecular exchange leads to a recovery of MR-visible water molecules in the blood regions and the ADC’ approaches the equilibrium ADC. Relaxation back to the equilibrium ADC can be described by the apparent exchange rate constant, AXR.
  • Directions or Averages? An Ablation Study for in vivo Cardiac DTI
    Jaume Coll-Font1,2,3, Shi Chen1, Robert A Eder1, and Christopher T. Nguyen1,2,3
    1Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Charlestown, MA, United States, 2Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, United States, 3Harvard Medical School, Boston, MA, United States
    We evaluated the differences in gradient direction acquisition schemes for whole-heart cardiac DTI and demonstrated that acquiring DW images along different gradient directions is marginally preferable to acquiring multiple repetitions of the same directions.
    Figure 1. Parameter maps of a representative subject obtained for the different combination of gradient orientations. The rows correspond to the results for all acquired gradient directions (reference: 30 dir. x2), 30x1, 15x2 and 15x1 directions. The maps obtained with at least 30 samples resulted were similar. On the other hand, the DTI fits on only 15 directions resulted in overestimation of FA and disrupted HA maps.
    Figure 2. DTI parameters projected on the AHA segments and averaged for all subjects. MD and FA are most robust to different gradient direction selection approaches. On the other, FA and HAT are sensitive to a reduction in the number of samples, as can be observed in the differences of the results obtained with 15 Dir. x1.
  • Time-dependent anisotropic diffusion in the mouse heart: feasibility of motion compensated tensor-valued encoding on a 7T preclinical scanner
    Samo Lasic1,2, Henrik Lundell1, Beata Wereszczyńska3, Matthew Budde4, Nadira Yuldasheva3, Filip Szczepankiewicz5, Erica Dall’Armellina3, Jürgen E. Schneider3, and Irvin Teh3
    1Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Hvidovre, Copenhagen, Denmark, 2Random Walk Imaging, Lund, Sweden, 3Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom, 4Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, United States, 5Clinical Sciences, Lund University, Lund, Sweden
    Acceleration compensated tensor-valued encoding at high b-values is feasible on a preclinical scanner. Time-dependent diffusion was observed in mouse hearts.
    Figure 1: Effective gradient waveforms g(t), dephasing waveforms q(t), normalized spectral trace s(ω) and diagonal components of the dephasing cross power spectral density. Individual gradient components (XYZ shown in red, green, blue) from the motion compensated STE waveforms (M0-, M1-, M2-STE) were used as M0-, M1-, M2-LTE. with all moments nulled up to zeroth, first and second order (M0, M1, M2). Their power spectra is shown below s(ω). Note the shift of spectra from zero frequency to higher frequencies, which alters sensitivity to time-dependent diffusion.
    Figure 2: ROI-average mean diffusivity in the left ventricle myocardium in hearts 1 (top) and 2 (bottom) and theoretical prediction. The ROIs are shown the right. Left column: measured MD (markers) and prediction (lines) vs cylinder radius, R. Right column: measured MD vs prediction with error bars corresponding to the distance between measurement and prediction (dotted line for unity relation). The n=1,2,3 labels for Mn-LTE correspond to the X,Y,Z channels of Mn-STE.
  • Evaluation of Synthetic-DWI with T2-based Water Suppression for DTI
    Tokunori Kimura1, Kousuke Yamashita1, and Kouta Fukatsu1
    1Department of Radiological Science, Shizuoka College of Medicalcare Science, Hamamatsu, Japan
    We clarified that our proposed T2wsup-DWI technique was superior to already proposed water suppression DWI methods of FLAIR and non-b-zero (NZE) methods in both of the ADC-SNR and the reduction effects of CSF partial volume effects (PVE) in DTI parameters of ADC, FA, and fiber tractograpy.
    Fig. 4. Results of MR brain study Comparison of DTI images, quantitative maps, and fiber tractographies, for standard (std) and for our water suppression (T2wsup). The T2wsup provided better tissue-specific values for the tissues close to the boundary regions of the ventricle, as the body of fornix (orange arrows), or genu of corpus callosum (yellow arrows). For the tractographies of fornix, the fibers at the central portions of two seed ROIs (yellow arrow) for T2wsup was thicker and better connected than for the standard, and artifactual fibers were drawn for the standard (blue arrow).
    Fig. 3. Simulation results with 2-point method. A: MD and FA as a function of Vw (%) for standard SE-DWI (std) and T2wsupSE-DWI (T2wsup) obtained by the two-point method with denoted combinations of b = (b0, bn) [s/mm2]. B: SIs as a function of b-value (b) for three DWI methods (a–c) each as a parameter of water volume ratio (Vw), and overlapped version a and b (d). C: Theoretical SNRs of ADC values as a function of the second b-value, bn of pure tissue (Vw = 0, ADC = 0.8×10−3 mm2/s) for three DWI techniques with two-point method when the SNR of SE b0 image (S(b0)/σ) = 50.
  • Toward high-resolution mapping of microscopic anisotropy in the cortex using b-tensor diffusion imaging with a spiral readout at 7 Tesla
    Sajjad Feizollah1,2 and Christine L. Tardif1,2,3
    1Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada, 2McConnell Brain Imaging Center, Montreal Neurological Institute, McGill University, Montreal, QC, Canada, 3Department of Biomedical Engineering, McGill University, Montreal, QC, Canada
    A b-tensor diffusion imaging sequence with a spiral readout was implemented at 7T with dynamic field monitoring. We present maps of the microscopic anisotropy of the brain at 1.4 mm isotropic with minimal geometric distortions.
    Figure 4- Mean diffusivity (MD), fractional anisotropy (FA), and microscopic anisotropy (µFA) maps for 3 transverse slices.
    Figure 3- Reconstructed images of 3 slices. The first column on the left shows images acquired by the Cartesian GRE sequence for comparison. Other columns show reconstructed STE images for 4 b-values with the same diffusion direction. Images of each b-value are presented with a different scale for better visibility. The yellow arrow shows remaining artifacts caused by static field inhomogeneities in the frontal lobe.
  • Reducing Rician noise bias in axial-symmetric Diffusion Kurtosis Imaging and biophysical tissue models
    Jan Malte Oeschger1, Karsten Tabelow2, and Siawoosh Mohammadi1,3
    1Institute of Systems Neuroscience, University Medical Center Hamburg-Eppendorf, Hamburg, Germany, 2Weierstrass Institute for Applied Analysis and Stochastics, Berlin, Germany, 3Department of Neurophysics, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
    The reduced axial symmetric DKI model is well suited to estimate DKI and biophysical tissue parameters. Combination with Rician noise bias correction improves parameter estimation, especially for DKI parameters along the main fiber direction.
    Figure 1: Top: In-vivo DWI images and white matter regions of interest (ROI) identified with the Jülich fiber atlas10, a) optic radiation (or), b) cortico spinal tract (ct), c) superior longitudinal fasciculs (slf) and d) callosum body (cb). In every one of the four ROIs, three voxels were chosen (FA images bottom, for slf only one is shown here) and DKI parameters were estimated with the standard DKI model without RBC. These 12 sets of DKI parameters were then used to generate ground truth signals for the simulation.
    Figure 2: Bias and standard deviation for the estimated AxDKI model parameters, averaged over 2500 noise samples per SNR across all ROIs. Note the differently scaled y axes for the diffusion (top) and kurtosis parameters (bottom). The axial-symmetric fit with RBC (magenta line) produces the overall least biased estimators.
  • Cerebrospinal Fluid Partial Volume Effects in Microscopic Fractional Anisotropy Imaging
    Nico J. J. Arezza1,2 and Corey A. Baron1,2
    1Medical Biophysics, Western University, London, ON, Canada, 2Centre for Functional and Metabolic Mapping, Robarts Research Institute, London, ON, Canada
    We investigated two techniques to remove free-water partial volume effects in microscopic fractional anisotropy (μFA) imaging. The techniques outperformed standard diffusion imaging methods in simulations and an expected increase in μFA and decrease in diffusivity were observed in vivo.
    Figure 3. Example in vivo mean diffusivity (top) and microscopic fractional anisotropy (bottom) slices estimated using the DTI model (left), DKI model (center-left), shifted DKI model (center-right), and FWE-DTI model (right) with data from STE acquisitions.
    Figure 1. Summary of the models used to estimate diffusivity from STE signal data in voxels containing CSF and tissue: (a) DTI, (b) DKI, (c) FWE-DTI (with separate compartments shown by dashed lines), and (d) shifted DKI. The C terms represent constants. In FWE-DTI, signal due to CSF-PVEs is removed by separating the overall signal into diffusion components for free water and brain tissue. In shifted DKI, signal due to CSF contamination is attenuated via the use of higher b-values.
  • Development of in vivo human brain DTI-MRE: Optimization of experimental parameters
    Shujun Lin1, Bradley Sutton2, Richard Magin1, Aaron Anderson2, and Dieter Klatt1
    1Richard and Loan Hill Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, United States, 2Beckman Institute, University of Illinois at Urbana-Champaign, Urbana-Champaign, IL, United States
    Our experiments confirm optimized experimental parameters identified in simulations.
    Figure 3. Frequency-dependent stiffness maps of a central slice from both DTI-MRE and conventional MRE.
    Figure 4. Mean diffusivity and fractional anisotropy maps of the same central slice from DTI-MRE and conventional DTI excluding fluid-filled ventricles.
Back to Top
Digital Poster Session - Diffusion Applications: Cancer
Diffusion/Perfusion
Thursday, 20 May 2021 13:00 - 14:00
  • Correcting B0 inhomogeneity-induced distortions in whole-body diffusion MRI of bone metastases
    Leonardino A. Digma1, Christine H. Feng1, Christopher C. Conlin2, Ana E. Rodriguez-Soto2, Kanha Batra3, Aaron Simon1, Roshan Karunamuni1, Joshua Kuperman2, Rebecca Rakow-Penner2, Michael E. Hahn2, Anders M. Dale2, and Tyler M. Seibert1,4
    1Department of Radiation Medicine and Applied Sciences, UCSD School of Medicine, La Jolla, CA, United States, 2Department of Radiology, UCSD School of Medicine, La Jolla, CA, United States, 3Department of Electrical and Computer Engineering, UC San Diego, La Jolla, CA, United States, 4Department of Bioengineering, UC San Diego, La Jolla, CA, United States
    In this work, we demonstrate that DWI of bone metastases undergo spatial distortions due to static magnetic field inhomogeneities. We also show that these distortions can be efficiently corrected using the reverse polarity gradient technique.
    Figure 1. Example bone metastasis illustrating B0 inhomogeneity induced distortion on DWI. (A) Pre-DisCo DWI b=2000 s/mm2 (B) Lesion annotation (pink) overlaid on post-DisCo DWI b=2000 s/mm2 (C) Distortion map; voxel values represent extent of displacement at each voxel. Red and blue values denote displacement in the posterior and anterior direction, respectively. (D) Pre-DisCo DWI b=0 s/mm2 (E) Post-DisCo DWI b=0 s/mm2. F. T2-weighted image. In each subfigure, bounding box was drawn 10 voxels from the lateral edge of the lesion. Abbreviations: DisCo=distortion correction.
    Figure 3. Distribution of RMS distortion and MI between b=0 s/mm2 and T2-weighted images. (A) RMS distortion for all lesions (black) as well as the distribution within specific imaging stations. (B) Change in MI values between the b=0 s/mm2 and T2 images after DisCo. A value larger than 0 indicates improved agreement between b=0 and T2 images. (C) RMS distortion for lesions broken down by anatomic group. (D) Change in MI values after DisCo by anatomic group. Abbreviations: RMS=root mean square, MI=mutual information.
  • Developing a multi-parametric model of response based on biomarkers derived from Whole-Body Diffusion Weighted Imaging
    Antonio Candito1, Matthew D Blackledge1, Fabio Zugni2, Richard Holbrey1, Sebastian Schäfer3, Matthew R Orton1, Ana Ribeiro4, Matthias Baumhauer3, Nina Tunariu1, and Dow-Mu Koh1
    1The Institute of Cancer Research, London, United Kingdom, 2IEO, European Institute of Oncology IRCCS, Milan, Italy, 3Mint Medical, Heidelberg, Germany, 4The Royal Marsden NHS Foundation Trust, London, United Kingdom
    Developing a multi-parametric model of response in patients with metastatic bone disease based on biomarkers derived from Whole-Body Diffusion Weighted Imaging. A change in median ADC showed significance in identifying response. The model predicted response with an accuracy of 87%.
    Table 1. Baseline characteristics for all the 41 patients. The blood-based biomarkers investigated were the Prostate-Specific Antigen (PSA), Hemoglobin level (HGB), Lactate dehydrogenase level (LDH) and Alkaline phosphatase level (AlkpH).
    Figure 1. A) Probability of response derived from the trained logistic regression model which expects as features the relative change of Median gADC and tDV. B) Probability of response and 95% confidence interval (dashed lines) derived from fitting the logistic regression model input only the relative change of Median gADC. Estimates of the probability of response higher than 60% and lower than 40% showed low uncertainty. However, samples with a probability of response from 40 to 60% (and around the decision boundaries as showed in Figure 1A) shown higher uncertainty.
  • Developing a deep learning model to classify normal bone and metastatic bone disease on Whole-Body Diffusion Weighted Imaging
    Antonio Candito1, Matthew D Blackledge1, Fabio Zugni2, Richard Holbrey1, Sebastian Schäfer3, Matthew R Orton1, Ana Ribeiro4, Matthias Baumhauer3, Nina Tunariu1, and Dow-Mu Koh1
    1The Institute of Cancer Research, London, United Kingdom, 2IEO, European Institute of Oncology IRCCS, Milan, Italy, 3Mint Medical, Heidelberg, Germany, 4The Royal Marsden NHS Foundation Trust, London, United Kingdom

    Developing a transfer learning model to classify normal bone and metastatic bone disease on Whole-Body Diffusion Weighted Imaging. The model shows promising results in classifying normal/metastatic bone on individual WBDWI slices, potentially without the requirement of Dixon imaging

     

    Figure 3. Assess model performance on 3 patients of the test data. Coronal and sagittal Maximum-Intensity-Projection (MIP) of SNRmap. The manual delineation of bone lesions performed by an experienced radiologist was overlayed on the MIP of SNRmap (in red). The first colormap shows the ground truth, annotated axial images with (in red) and without (in blue) metastatic bone disease. The second colormap shows the prediction performed by the trained transfer learning model.
    Figure 5. Axial Fat fraction, b900, RGB image and CAM from test patient 2 in Figure 3. Fat fraction combined with b900 images are typically used for manual detection of bone disease. However, using the DWI data alone, our model was able to predict presence of disease in one slice (top-row, red outline), and absence of disease in two different slice locations. CAM emphasizes where the model is ‘looking’ to derive the relevant regions within the images that lead to a particular decision. From the heatmap, red = important and blue = not important.
  • Improved assessment of prostate-cancer bone metastases through multicompartmental analysis of whole-body DWI data
    Christopher C Conlin1, Christine H Feng2, Leonardino A Digma2, Ana E Rodriguez-Soto1, Joshua M Kuperman1, Dominic Holland3, Rebecca Rakow-Penner1, Tyler M Seibert1,2,4, Michael E Hahn1, and Anders M Dale1,3,5
    1Department of Radiology, UC San Diego School of Medicine, La Jolla, CA, United States, 2Department of Radiation Medicine and Applied Sciences, UC San Diego School of Medicine, La Jolla, CA, United States, 3Department of Neurosciences, UC San Diego School of Medicine, La Jolla, CA, United States, 4Department of Bioengineering, UC San Diego Jacobs School of Engineering, La Jolla, CA, United States, 5Halıcıoğlu Data Science Institute, UC San Diego, La Jolla, CA, United States
    An optimized 4-compartment model better characterized whole-body diffusion than conventional DWI methods. Compartmental signal-contributions revealed by this model may help to detect and quantify prostate-cancer bone involvement.
    Figure 3: Coronal whole-body images of a patient with metastatic bone lesions in the pelvis and femur (red arrows). Conventional MR images are shown in the top row. The bottom row shows the signal-contribution (Ci) maps for the optimized 4-compartment RSI model. The corresponding Di of each model compartment is listed in parentheses next to the compartment label.
    Figure 2: Model-fitting residual at the voxel- and ROI-level. The top row shows voxel-wise maps of fitting residual in a coronal plane of the same patient using different models. A T2-weighted image of the same plane is included for reference. The bottom figure graphs the fitting residual within all lesion and tissue-specific ROIs. A better fit to the data was observed with the 4-compartment RSI model than with the conventional monoexponential model or the lower-order RSI models.
  • Assessment the Preponderant Diagnostic Performances of Oligometastatic Prostate Cancer Using Diffusion Kurtosis Imaging
    Suhong Qin1, Ailian Liu1, SHUANG MENG1, Lihua Chen1, Qinhe Zhang1, Qingwei Song1, and Yunsong Liu1
    1The First Affiliated Hospital of Dalian Medical University, Dalian, China
     The performances of DKI cannot differentiate between oligometastatic and widely metastatic PCa, but it has the potential to assess tumor load and aggressiveness in metastatic PCa.
    Figure 1 A 69-year-old male with PCa.ROI was manually placed on the local lesion of MK map (A). The Ka, Kr, FAk, MD, Da, Dr and FA maps of DKI were shown(B-H).

    Table 2 Results of correlation analysis of DKI parameters and PSA

    * P value is statistically significant.

  • Methodological considerations on segmenting MRI data of rhabdomyosarcoma
    Cyrano Chatziantoniou1,2, Reineke Schoot2, Roelof van Ewijk2, Simone ter Horst2, Rick van Rijn3, Hans Merks2, Alexander Leemans1, and Alberto de Luca1
    1Image Science Institute, UMC Utrecht, Utrecht, Netherlands, 2Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands, 3Department of Radiology, Academic Medical Centre Amsterdam, Amsterdam, Netherlands
    In surveying recent literature on sarcomas, we found that segmentation strategies vary strongly. In comparing these strategies on a toy example, we found that the choice of segmentation method can yield a difference in measured diffusion of up to 23%.
    Figure 1: Segmentation methods as used in 52 recent papers on sarcoma shows a large variety in important aspects of the segmentation strategy.
    Choice of the segmentation strategy can have large impact on mean and minimum ADC (apparent diffusion coefficient) measured over the segmented area. Segmentation performed by two experienced radiologists (rater 1 and 2, outline) and a trained researcher (circular and 3D) on an ADC map of pediatric rhabdomyosarcoma. Segmented areas are shown in figure 3.
  • Non-gaussian IVIM DW-and fast exchange regime DCE- MRI for predicting of locoregional failure in nasopharyngeal carcinoma
    Ramesh Paudyal1, Linda Chen2, Jung Hun Oh1, Kaveh Zakeri2, Vaios Hatzoglou3, Chiaojung Jillian Tsai2, Nancy Lee2, and Amita Shukla-Dave1,3
    1Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, United States, 2Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, United States, 3Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
    Quantitative imaging metrics derived from NG-IVIM DW- and FXR  DCE-MRI  could predict the patients with LRF in NPC  using the cumulative incidence analysis .
    Figure 4. Cumulative incidence analysis for pretreatment apparent diffusion coefficient, ADC (mm2/s), true diffusion coefficient, D (mm2/s), perfusion fraction, f, volume transfer constant, Ktrans (min-1), the volume fraction of extravascular extracellular, ve, and mean lifetime of intracellular water protons, τi (sec). Gray’s test revealed a significant difference for ADC, D, and f (P <0.05), and a borderline significance for τi (sec) (P=0.098).
    Figure 3. Left: Representative histogram plot of true diffusion coefficient, D, and kurtosis coefficient, K), in patients with and without LRF of nasopharyngeal cancer (Figure 2). D values spread out towards higher in a patient without LRF than compared to with LRF. In contrast, K values spread to a higher value in a patient with LRF than without LRF. Right: Ktrans values spread out towards higher in a patient without LRF as compared to with LRF. In contrast, τi values spread to higher in a patient with LRF as compared without LRF.
  • Repeatability of VERDICT diffusion MRI in a model of human neuroendocrine tumour
    Lukas Lundholm1, Mikael Montelius1, Oscar Jalnefjord1, Eva Forssell-Aronsson1, and Maria Ljungberg1
    1Department of Radiation Physics, Institute of Clinical Sciences, Gothenburg, Sweden
    VERDICT dMRI showed a good overall repeatability of the mean estimated parameter values within the studied tumours. However, some local clusters of voxels showed larger differences between repeated scans.
    Figure 2. Colormaps of cell radius index (R), intracellular volume fraction (fIC), vascular volume fraction (fVASC), and extracellular extravascular volume fraction (fEES) as estimated by VERDICT in the central slice of an example tumour. Colormaps are shown for two repeated measurements (M1 and M2) and the difference between the repeated measurements (Δ)
  • Qualitative and quantitative comparison between IVIM-DKI and PET/CT imaging in lymphoma
    Archana Vadiraj Malagi1, Devasenathipathy Kandasamy2, Kedar Khare3, Deepam Pushpam4, Rakesh Kumar5, Sameer Bakhshi4, and Amit Mehndiratta1,6
    1Centre for Biomedical Engineering, Indian Institute of Technology Delhi, New Delhi, India, 2Department of Radiodiagnosis, All India Institute of Medical Sciences Delhi, New Delhi, India, 3Department of Physics, Indian Institute of Technology Delhi, New Delhi, India, 4Department of Medical Oncology, Dr. B.R. Ambedkar Institute-Rotary Cancer Hospital (IRCH), All India Institute of Medical Sciences Delhi, New Delhi, India, 5Department of Nuclear Medicine, All India Institute of Medical Sciences Delhi, New Delhi, India, 6Department of Biomedical Engineering, All India Institute of Medical Sciences Delhi, New Delhi, India
    Correlation analysis showed no significant relationship between IVIM-DKI and PET parameters. IVIM parameters with Total-Variation model produced substantial reproducibility with better quality parameter maps.
    Figure2 (a) SUV map, (b) ADC map, (c, k) DKI parameter maps, (d, e) diffusion parameters and (g-j) perfusion parameters from IVIM analysis of a 32-year-old male was diagnosed for NHL; the tumour was present in sternum encircled in red.
    Figure1 PET images (red) overlapped onto ADC images of a patient with NHL, where the tumour is encircled (green). (a) to (g) represents baseline scans at axial view. (h) Representative 3D sagittal view with lines indicating slices (a-g) selected for visualization.
  • Preliminary Study on Monitoring Drug Resistance of Colon Cancer with Intravoxel Incoherent Motion MRI In Vivo
    Qi Xie1, Wenjuan He1, Zhilin Tan1, Yajie Wang1, Jinbin Wu1, Xiyan Shao2, Yiming Yang3, Jing Zhang4, Kangwei Wang5, Guiqin Wang6, Qifeng Pan1, and Yunzhu Wu7
    1Medical Imaging Department, Nansha Hospital, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China, 2Ultrasound Imaging Department, Longgang District People’s Hospital, Shenzhen, China, 3Department of Radiology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou, China, 4Department of Pathology, Cancer Center, Sun Yat-sen University, Guangzhou, China, 5Department of Pathology, Nansha Hospital, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China, 6Medical Record Department, Nansha Hospital, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China, 7MR Scientific Marketing, SIEMENS Healthcare Ltd., Guangzhou, China
    The ADC value of DWI and diffusion coefficient D of IVIM were moderate valuable for discriminating 5-FU-response and resistance of colon cancer in vivo.
    IVIM DWI parametric maps of ADC, D, D* and f values and T2WI images for SW480 (A-E)and SW480/5-FU(F-J).
    The Comparison of The ADC(a), D(b), D*(c) and f(d) values between SW480/5-FU and SW480 xenografts.
  • Evaluation of bone marrow infiltration in the newly diagnostic Multiple Myeloma with Intravoxel Incoherent Motion Diffusion-weighted MRI
    Xiaojiao Pei1, Tao Jiang1, Zhenyu Pan1, Yufei Lian1, Yueluan jiang2, and qinglei Shi 3
    1Radiology, Beijing Chaoyang Hospital Affiliated to Capital Medical University, Beijing, China, 2MR Scientific Marketing, Diagnosis Imaging, Siemens Healthineers China, Beijing, China, 3Scientific Marketing, Diagnosis Imaging, Siemens Healthineers China, Beijing, China
    This study used IVIM parameters to quantitatively diagnose MM vertebral bone marrow and normal bone marrow. For diagnosing MM patients from healthy volunteers, the D showed the highest sensitivity was 90.0%, and its Specificity was 94.7%,with the cutoff value of 141.8×10-3㎜2/s.
    Fig1. Exemplary Sagittal IVIM parametric maps of lumbar spine in NDMM group and healthy group.
    Fig3. Analysis of ROC curve between ADC, D and D*. The area under the receiver operating characteristic (ROC) curve of ADC, D and D* were 0.935± 0.037, 0.944± 0.034, and 0.935± 0.036, respectively. Among these, the IVIM-dfast showed the highest sensitivity was 90.0%, and its Specificity was 94.7%, with the cutoff value of 141.8×10-3㎜2/s.
  • Diffusion and perfusion MRI predicts response preceding and shortly after stereotactic radiosurgery to brain metastases
    Amaresha Shridhar Konar1, Akash Deelip Shah2, Ramesh Paudyal1, Jung Hun Oh1, Eve LoCastro1, David Aramburu Nuñez1, Nathaniel Swinburne2, Robert J. Young2, Andrei I. Holodny2, Kathryn Beal3, Vaios Hatzoglou2, and Amita Shukla-Dave1,2
    1Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, United States, 2Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, United States, 3Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
    The present prospective study aims to determine the ability of DW- and DCE-MRI to predict the long-term response of Brain Metastases within 72 hours of SRS. The results are promising as it will inform the treating physicians at an early time point about which patients will benefit from SRS (or not).
    Figure 1: Representative pre-SRS and post-SRS (within 72 hours of treatment) MR images from an 80 year-old female with right occipital lobe brain metastasis, which demonstrated progressive disease (PD). Top panel: D(×10-3 mm2/s), D*(×10-3 mm2/s), and f maps are zoomed in at the locations of ROIs (pre-SRS b=0 s/mm2 image). The lesion volume was unchanged at post-SRS. Bottom panel: Ktrans (min-1), ve, and vp maps are zoomed in at the locations of ROIs (pre-SRS T1-postcontrast image).
    Figure 4: Lesions that progressed had a higher f mean and median than those that did not. Lesions that responded to SRS had a lower pre-SRS Ktrans mean, Ktrans median, and ve median than those that did not. ADC mean and median trended higher in lesions post-SRS than pre-SRS. vp was lower in lesions post-SRS than pre-SRS. ADC, apparent diffusion coefficient. Ktrans, index of tumor vascular permeability. ve, volume fraction of extracellular extravascular space. vp, vascular volume.
  • The utility of IVIM maps in the assessment of microvascular perfusion of brain glioma
    Andre Monteiro Paschoal1,2, Raquel Andrade Moreno3,4, Antonio Carlos dos Santos5, and Renata Ferranti Leoni6
    1LIM44, Instituto e Departamento de Radiologia, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil, 2InBrain Lab, University of Sao Paulo, Ribeirao Preto, Brazil, 3Instituto do Cancer do Estado de Sao Paulo, Sao Paulo, Brazil, 4Memorial Sloan-Kettering Cancer Center, New York, NY, United States, 5Departamento de Clinica Medica, Faculdade de Medicina de Ribeirao Preto, Universidade de Sao Paulo, Ribeirao Preto, Brazil, 6InBrain Lab - University of Sao Paulo, Ribeirao Preto, Brazil
    IVIM fD* maps were able to detect small perfusion changes in early stages of low grade gliomas, suggesting its use as an early biomarker for this parameter. In high grade glioma, it may provide complementary information to contrast agent perfusion.
    Figure 1: High grade glioma. a) DSC CBF map; b) IVIM fD* map; c) SWI map; d) Correlation between fD* and CBF at the tumor ROI in yellow at the tumor border; e) Correlation between fD* and CBF at the tumor ROI in green at the tumor interior.
    Figure 2: Low grade glioma. a) DSC CBF map; b) DSC CBV map; c) IVIM fD* map; d) FLAIR; e) Post contrast T1 image; f) SWI; g) Correlation between fD* and CBF at the tumor ROI at the tumor border; h) Correlation between fD* and CBF at the tumor ROI at the tumor interior.
  • Diffusion-time-dependent diffusion MRI based microstructural mapping for grading and categorizing in pediatric brain tumor
    ruicheng ba1, Hongxi Zhang2, Zhongwei Gu3, Yuhao Liao1, Xingwang Yong1, Zhiyong Zhao1, Yi Zhang1, and Dan Wu1
    1Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering & Instrument Science, Zhejiang University, Hang zhou, Zhejiang, China, 2Children's Hospital, Zhejiang University School of Medicne,Department of Radiology, Hang zhou, Zhejiang, China, 3Children's Hospital, Zhejiang University School of Medicne, Department of Pathology, Hangzhou, Zhejiang, China
    Tumor microstructure was estimated based on time-dependent dMRI and IMPULSED model. Cellularity and intracellular fraction increased from low-grade to high-grade glioma, and further increased in medulobastom. Cellularity achieved the highest area-under-the-curve in grading  glioma.
    Figure 1: Gd-enhanced T1w images and the maps of ADC(OGSE-17Hz, OGSE-33Hz, PGSE), cellularity, intracelluar fraction and cell diameter as obtained from the IMPULSED model. The ADC and microstructural maps were overlaid on the b0 image for reprehensive low-grade glioma, high-grade glioma, and medulloblastoma cases.
    Figure 4: (A) Correlation between ADC (PGSE) and cellularity. Regression analysis revealed correlation was strongest for low-grade glioma (r = -0.8623, p = 0.0003), followed by high-grade glioma (r = -0.7246, p = 0.0117); whereas not significant correlation were found for medulloblastoma (p=0.4095). (B) Similar correlations patterns were found between ADC (PGSE) and Intracellular fraction.
  • Imaging attributes of H3K27M mutation in Diffuse Midline Gliomas on Multiparametric MRI
    Richa Singh Chauhan1, Nihar Kathrani2, Jitender Saini1, Maya D Bhat1, Karthik Kulanthaivelu1, Vani Santosh3, Nishanth S4, and Subhas Konar4
    1Neuroimaging and Interventional Radiology, NIMHANS, BENGALURU, India, 2Interventional Radiology, Paras Hospital, Gurgaon, India, 3Neuropathology, NIMHANS, BENGALURU, India, 4Neurosurgery, NIMHANS, BENGALURU, India
    H3K27M mutation in diffuse midline gliomas can be predicted on multiparametric MRI using the conventional and advanced (diffusion and perfusion) sequences.
    (A to I) T1w, T2w, FLAIR, DWI, ADC, SWI, post-contrast T1w images, rCBV map, and perfusion curve of a bithalamic H3K27M mutant DMG.
    (A to I) T1w, T2w, FLAIR, DWI, ADC, SWI, post-contrast T1w images, rCBV map, and perfusion curve of a left thalamic H3K27M wild-type DMG.
  • Grading of glioma with histogram analysis of multiparameter using advanced diffusion models
    Gao Eryuan1, Gao Ankang1, Zhang Huiting2, Wang Shaoyu2, Yan Xu2, Bai Jie1, and Cheng Jingliang1
    1Dept. of MRI, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China, Zhengzhou, China, 2MR Scientific Marketing, Siemens Healthcare, Shanghai, China, Shanghai, China
    Histogram Analysis based on multiple diffusion models was helpful in glioma grading., especially for the maximum values of DTI and MAP methods.
    Fig. 1. Bar graphs of ADmaximum, MDmaximum, RDmaximum, QIVmaximum and QIVrange values averaged across grade Ⅱ (n=33), grade Ⅲ (n=11) and grade Ⅳ (n=54) gliomas. All parameters are significant with P<0.05.
    Fig. 2. ROC analysis for ADmaximum, MDmaximum, RDmaximum, QIVmaximum and QIVrange in three comparisons (GradeⅡ VS GradeⅢ, GradeⅡVS Grade Ⅳ and GradeⅢ VS Grade Ⅳ)
  • Histogram analysis in prediction of Isocitrate Dehydrogenase Genotype in Gliomas with MRI: The Gaussian versus non-Gaussian Diffusion Models
    Gao Ankang1, Gao Eryuan1, Zhang Huiting2, Wang Shaoyu2, Yan Xu2, Bai Jie1, and Cheng Jingliang1
    1Dept. of MRI, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China, 2MR Scientific Marketing, Siemens Healthcare, Shanghai, China, Shanghai, China
    MAP as an advanced diffusion model has more advantages than other models in prediction of glioma Genotype. Histogram analysis is a useful method in quantitative analysis of diffusion parameters and shows a great potential in glioma research.
    The distribution of mean and SD of useful parameters
    The list for the parameters of diffusion models and histogram analysis
  • Neurite Orientation Dispersion and Density Imaging in Evaluation of Glioma-induced Corticospinal Tract Injury
    Rifeng Jiang1, Kaiji Deng1, Yixin Guo2, and Zhongshuai Zhang3
    1Fujian Medical University Union Hospital, Fuzhou, China, 2Fujian Medical University, Fuzhou, China, 3MR Scientific Marketing, Siemens Healthcare, Shanghai, China
    This study found that NODDI seems to be a more potent approach in evaluating the early CST infiltration by HGG, and can evaluate the CST destruction with a similar performance to MD by providing additional information about neurite density for HGG-induced CST injury.
    Three representative subjects demonstrating the CST changes in HGG patients. The subject A was a 39-year-old healthy male. All the CST features have good bilateral symmetry. The patient B was a 72-year-old male with glioblastoma, and his motor function was normal. The ICVF of the affected CST decreased obviously, but changes of the affected CST were not obvious in the other diffusion parameters. The patient C was a 62-year-old female with glioblastoma, and she had motor weakness (grade 2). Changes of the affected CST were obvious in FA, MD and ICVF, but not in ISOVF and ODI.

    Changes of relative CST features in HGG patients. Box and whisker plot (A-C) showed that compared with the relative CST features in HGG_NMF, the relative MD was significantly higher, where the relative FA and ICVF were significantly lower (*) in HGG_MW; compared with the relative CST features in HC, the relative ICVF was significantly lower (*) in HGG_NMF; in contrast, no significant changes were found in the relative CST morphological characteristics, ISOVF or ODI.

    HGG_MW = HGG patients with motor weakness, HGG_NMF = HGG patients with normal motor function, HC = healthy subject.

  • Beyond cellularity: Which microstructural features determine the mesoscopic mean diffusivity in meningiomas?
    Jan Brabec1, Filip Szczepankiewicz2, Jaromír Šrámek3, Elisabet Englund4, Johan Bengzon5, Linda Knutsson1,6, Carl-Fredrik Westin7,8, Pia C Sundgren2,9, and Markus Nilsson2
    1Medical Radiation Physics, Lund University, Lund, Sweden, 2Diagnostic Radiology, Lund University, Lund, Sweden, 3Institute of Histology and Embryology, First Faculty of Medicine, Charles University, Prague, Czech Republic, 4Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden, 5Division of Neurosurgery, Department of Clinical Sciences, Lund University, Lund, Sweden, 6Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States, 7Harvard Medical School, Boston, MA, United States, 8Radiology, Brigham and Women’s Hospital, Boston, MA, United States, 9Lund University Bioimaging Center, Lund University, Lund, Sweden
    Cellularity poorly explains intra-tumor variability of mean diffusivity on mesoscopic scale in meningiomas. Other microstructural features such as microcysts, macrocysts, vessels, psammoma bodies, collagen fibers or need to be considered.
    Figure 1. Analysis of microcystic/angiomatous meningioma. The upper row shows measured (A) and predicted mean diffusivity based on cellularity (B): a second-degree polynomial yielded R2 of 0.19 (C). The error map (D) shows large negative (green area) and positive (red) errors where the predicted diffusivity is over/under-estimated. Overestimation was related to high prevalence of microcysts (a; bottom row) because these provide restrictions. Underestimation was found in regions that contained macrocysts and vessels (b and c) because these provide free water compartments.
    Figure 3. Quantitative analysis. Part A shows predicted diffusivity omitting and part B including microcysts. The error maps reveal large systematic error areas (yellow arrows) that disappears when cysts are included in the fit already as an additional linear term. In addition, R2 increases from 0.19 to 0.41. The blue part marks newly emerged error area that was not previously there. Part C highlights the changes in the error maps. The changes are strongest in the middle part where the cyst density is also the highest. The changes overall strongly correlate with cyst density (R2 = 0.81).
  • Diffusion MRI based on a gamma distribution model for the differentiation of primary central nervous system lymphomas and glioblastomas
    Osamu Togao1, Akio Hiwatashi2, Toru Chikui3, Kazufumi Kikuchi2, Yukiko Kami3, Kenji Tokumori4, and Kousei Ishigami2
    1Molecular Imaging & Diagnosis, Kyushu University, Fukuoka, Japan, 2Clinical Radiology, Kyushu University, Fukuoka, Japan, 3Oral and Maxillofacial Radiology, Kyushu University, Fukuoka, Japan, 4Clinical Radiology, Teikyo University, Omuta, Japan
    The gamma distribution (GD) model analysis revealed that the κ, f2, and f3 values were smaller and the f1 values were larger in the primary CNS lymphomas than in the glioblastomas. The GD model may contribute to the characterization of brain tumor from the histological viewpoint.
    Figure 2. A 62-year-old-male with a PCNSL. A: The post-contrast T1-weighted image shows a ring-like enhancing mass lesion in the right frontal lobe (arrow). The enhancing lesion shows high signal intensity on the DWI (B) and a low ADC (0.70×10−3 mm2/sec, C). This lesion shows a small κ (1.76, D), a large θ (4.85×10−6 mm2/sec, E), a large f1 (0.626, F), a small f2 (0.270, G), and a small f3 (0.104, H).
    Figure 1. Comparisons of the GD model-derived parameters between the PCNSLs and GBs in the gadolinium-enhancing lesion. A: The κ was significantly smaller in the PCNSL group than in the GB group. B: The θ was not significantly different between the groups. C–E: The f1 was significantly larger and the f2 and f3 were significantly smaller in the PCNSL group than in the GB group.