Cancer: Contrast Agents & MRS
Cancer/Spectroscopy/Molecular Imaging/Pre-Clinical Monday, 17 May 2021
Digital Poster
939 - 958

Oral Session - Cancer: Contrast Agents & MRS
Cancer/Spectroscopy/Molecular Imaging/Pre-Clinical
Monday, 17 May 2021 12:00 - 14:00
  • High-resolution T1 Mapping of High-grade Glioma
    Zhibo Zhu1, Jay Acharya2, Yannick Bliesener1, R. Marc Lebel3,4, Richard Frayne3,5, and Krishna S. Nayak1,2
    1Ming Hsieh Department of Electrical and Computer Engineering, University of Southern California, Los Angeles, CA, United States, 2Department of Radiology, University of Southern California, Los Angeles, CA, United States, 3Radiology and Clinical Neuroscience, Hotchkiss Brain Institute, University of Calgary, Calgray, AB, Canada, 4Global MR Applications & Workflow, GE Healthcare, Calgary, AB, Canada, 5Seaman Family MR Research Centre, Foothills Medical Centre, Calgary, AB, Canada
    We evaluated a novel millimeter resolution whole-brain T1 mapping method in patient with high-grade glioma. Glioma T1 value is found to be larger and more heterogeneous compared to normal appearing white matter.
    Figure 1: M0 (left) and T1 (right) maps for three representative patients (F71, F61, F67) at the first time point. Maps are volumetric, and axial, coronal, and sagittal slices centered on the tumor section are shown. Tumor ROIs are delineated by red contours on M0 maps. Longer T1 can be observed in corresponding regions on the T1 maps. In addition, T1 maps reveal the locations of surgical cavities and extra-axial fluid collection (blue arrows) and evidence of craniotomy (green arrows). White dashed-line boxes indicate the zoomed-in region in Figure 2.
    Figure 2: Closeup of T1 maps from the axial, coronal and sagittal views on three patients shown in Figure 1. Maps are zoomed into the tumor region (delineated by white dashed lines in Figure 1), with constrained display range. The proposed method captures T1 heterogeneity. Light green: high T1 values, green: medium T1 values and dark green: low T1 values. Coefficients of variation for T1 inside the ROIs are 10.84%, 9.96%, and 7.31% for the top, middle, and bottom rows, respectively.
  • A nomogram combining T2WI-based radiomics features and clinical variables for prediction of neoadjuvant chemotherapy response in osteosarcoma
    Chengxiu Zhang1, Jingyu Zhong2, Yangfan Hu3, Jing Zhang1, Liping Si2, Yue Xing2, Jia Geng3, Qiong Jiao4, Huizhen Zhang4, Weiwu Yao2, and Guang Yang1
    1Shanghai Key Laboratory of Magnetic Resonance, East China Normal University, Shanghai, China, 2Department of Imaging, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China, 3Department of Radiology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China, 4Department of Pathology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
    A nomogram combined radiomics features from routinely available T2WI images and clinical variables to predict the response to neoadjuvant chemotherapy in patients with osteosarcoma was constructed and achieved an AUC of 0.838 (95% CI, 0.700-0.958).
    Fig. 1 Study workflow. 144 patients with osteosarcoma were randomly split into training (n = 101) and test (n = 43) dataset. Images were segmented manually. Radiomics features were extracted with Pyradiomics and data in training dataset were balanced with upsampling. Radiomics model building was performed with Pearson Correlated Coefficient for dimension reduction, RFE or Relief for feature selection, and SVM or LR for classifier. The final model was evaluated using ROC analysis, radiomics score plot, calibration curve, and decision curve analysis.
    Fig. 2 Clinical-radiomics combined nomogram and its utility. A nomogram (a) integrated the radiomics score and three clinical variables was constructed with an AUC of 0.838 in test dataset (b). The probability of pGR for each patient in terms of the response status in the training (c) and test dataset (d) suggested a good accuracy in response prediction. Decision curve analysis curves for clinical, radiomics and combined model in the whole dataset showed that the nomogram had a favorable clinical utility (e).
  • Using MR Radiomics to Improve Prediction of Local Tumor Control after Radiosurgery in Brain Metastases
    Chien-Yi Liao1, Cheng-Chia Lee2,3,4, Huai-Che Yang2,3, Wen-Yuh Chung2,3, Hsiu-Mei Wu3,5, Wan-Yuo Guo3,5, Ren-Shyan Liu1,6,7, and Chia-Feng Lu1,8
    1Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan, Taipei, Taiwan, 2Department of Neurosurgery, Neurological Institute, Taipei Veteran General Hospital, Taipei, Taiwan, Taipei, Taiwan, 3School of Medicine, National Yang-Ming University, Taipei, Taiwan, Taipei, Taiwan, 4Brain Research Center, National Yang-Ming University, Taipei, Taiwan, Taipei, Taiwan, 5Department of Radiology, Taipei Veteran General Hospital, Taipei, Taiwan, Taipei, Taiwan, 6Department of Medical Imaging, Cheng-Hsin General Hospital, Taipei, Taiwan, Taipei, Taiwan, 7Molecular and Genetic Imaging Core, Taiwan Animal Consortium, Taipei, Taiwan, Taipei, Taiwan, 8Institute of Biophotonics, National Yang-Ming University, Taipei, Taiwan, Taipei, Taiwan
    The prediction of treatment response after Gamma Knife stereotactic radiosurgery (GKRS) can benefit patient management. We suggested that imaging characteristics extracted from preradiosurgical MRIs combined with clinical information can effectively predict local tumor control.

    Figure 1. Image processing steps and radiomics flowchart

    (a) Image acquisition of T1cwc, T1w and T2w images, registration of T1w and T2w to T1c images and intensity normalization. (b) ROI delineation for the treatment planning of GKRS by experienced neurosurgeons and radiologists on all MRI slices covering tumor regions. (c) Extraction of radiomic features from the tumor ROIs on the MRIs.

    Figure 2. Representative cases and the model performance in predicting local tumor control

    (a) T1c images and reconstructed 3D tumor models. (b) The SVM scores in two representative cases based on the combination of radiomic and clinical features. (c) Receiver operating characteristic curves and the area under the curves (AUCs) of three SVM classification models.

  • The power of field strength: a direct comparison of USPIO-enhanced MRI at 3 and 7T to detect suspicious lymph nodes in patients with prostate cancer
    Ansje Fortuin1,2, Sjaak van Asten1, Andor Veltien1, Bart Philips1, Thomas Hambrock1, Stephan Orzada3,4, Harald Quick3,5, Jelle Barentsz1, Marnix Maas1, and Tom Scheenen1,3
    1Radboudumc, Nijmegen, Netherlands, 2Radiology, Ziekenhuis Gelderse Vallei, Ede, Netherlands, 3Erwin L Hahn Institute for MR Imaging, Essen, Germany, 4University of Heidelberg, Heidelberg, Germany, 5University of Duisburg-Essen, Essen, Germany
    USPIO-enhanced MRI in 20 patients with high-risk prostate cancer identified significantly more suspicious lymph nodes at 7T compared to 3T. Although annotating lymph nodes in the pelvis is not an easy task, 7T improves the interobserver agreement in scoring suspicious nodes.
    Table 2. Confusion matrices for reader scoring agreements on 99 from 410 co-identified nodes at 3T and on 159 from 601 co-identified nodes at 7T.
    Figure 2. Histogram of the size of annotated nodes by Reader A, Reader T and concordant Reader A & T for both 3 and 7 Tesla USPIO-enhanced MRI of patients with prostate cancer.
  • Radiomics-based CEST image analysis for improved performance of brain tumor grading
    Jibin Tang1, Hongxi Zhang2, Zhipeng Shen3, Wenqi Wang1, Xingwang Yong1, Junjie Wen1, Xinchun Chen2, Fengyu Tian2, Weibo Chen4, Dan Wu1, and Yi Zhang1
    1Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, Zhejiang, China, 2Department of Radiology, Children’s Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China, 3Department of Neurosurgery, Children’s Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China, 4Philips Healthcare, Shanghai, China
    We implemented a radiomic analysis of the APTw images, and found that the average sensitivity and AUC of the selected radiomic feature models for brain tumor grading were significantly higher than that of conventional mean APTw image intensities.
    Figure 1. Flowchart of the whole process.
    Figure 3. a: The average ROC curves of a representative 5-fold stratified cross-validation from radiomic analysis (red line) and coventional mean APTw signals (blue line). b: The pooled ROC curves (mean ± standard deviation) from 100 runs of the 5-fold stratified cross-validation, in which the red curves referred to results from radiomic analysis and blue curves denoted results from conventional mean APTw signals.
  • Delayed mapping of 2H-labeled choline using Deuterium Metabolic Imaging (DMI) reveals active choline metabolism in rat glioblastoma.
    Henk M. De Feyter1, Monique A. Thomas1, Kevan L. Ip1, Kevin L. Behar2, and Robin A. de Graaf3,4
    1Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, United States, 2Department of Psychiatry, Yale University, New Haven, CT, United States, 3Department of Radiology and Biomedical Imaging, Yale University, NEW HAVEN, CT, United States, 4Department of Biomedical Engineering, Yale University, New Haven, CT, United States
    After in vivo mapping of 2H-choline uptake with DMI, 2H NMR identifies choline species in metabolite extracts from rat glioblastoma, acutely and 24 hrs after infusion. The data show active metabolism of blood-borne choline in rat glioblastoma, and long retention of 2H in choline metabolites.
    Figure 2. DMI of [2H9]-Cho in rats with RG2 glioma. A, B) Contrast-enhanced T1w MRI, C-G) DMI maps acquired during a ~36 min IV infusion of [2H9]-Cho, showing the high signal in the tumor lesion in contrast to the surrounding normal brain. D-H), DMI maps acquired 20-24 hrs after a ~36 min IV infusion of [2H9]-Cho; note that panel A-D are from the same animal scanned on consecutive days. DMI amplitude based on peak integral of tCho signal, in a.u., and normalized to the highest value. I, J) 2H MR spectra from a voxel in the tumor, acquired during and after 2H-choline infusion, respectively.
    Figure 3. High resolution 2H NMR. Spectra acquired in metabolite extracts from excised tumor tissue, harvested immediately at the end (top), and 24 hrs after a 36 min infusion of [1,1,2,2-2H4]-Cho (bottom), Note the lack of free Cho after 24 hrs, indicating active metabolism of the blood-borne 2H-labeled Cho. Cho: choline, PC: phosphocholine, GPC, glycerophosphocholine. Note that spectra are from samples of different weights, and thus peak amplitudes are not necessarily quantitatively comparable.
  • Predictive Value of Myo-inositol Measured by MRSI during Anti-angiogenic Treatment in Recurrent Glioblastoma
    Michael Wenke1, Jorg Dietrich2, Elizabeth Gerstner2, Otto Rapalino3, Julian He3, Daniel Kim1, Melanie Fu1, Pratik Talati4, Mohamed El Abtah1, Anna Vaynrub1, Sharif Natheir1, Mark Vangel3, Isabel Arrillaga-Romany2, Forst Deborah2, Yi-Fen Yen1, Ovidiu Andronesi1, Jayashree Kalpathy-Cramer1, Tracy Batchelor5, Bruce Rosen1, R. Gilberto Gonzalez3, and Eva-Maria Ratai1
    1Radiology / Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, United States, 2Neurology / Cancer Center, Massachusetts General Hospital, Boston, MA, United States, 3Radiology, Massachusetts General Hospital, Boston, MA, United States, 4Neurosurgery, Massachusetts General Hospital, Boston, MA, United States, 5Neurology, Brigham and Women's Hospital, Boston, MA, United States
     Low tumoral myo-inositol prior to and during anti-angiogenic therapy is predictive of poor survival.

    Figure 2. Left: Representative MRS voxel selection on a T1WI post-contrast image, tumor voxels (red), peritumoral (green), and contralateral normal voxels (blue) were selected for analyses.

    Right: mI/c-Cr ratios at baseline in the tumor, (T) periphery (P), and contralateral (C) volumes of interest (VOI). Box plots indicate quartiles. Mean and standard deviations of mI/c-Cr in each VOI with t-tests indicating each VOI pair is significantly different.

    Figure 3. Mean and standard deviations of mI/c-Cr classified by OS9. the asterisk (*) indicate p <0.05.
  • Deuterium magnetic resonance spectroscopy using 2H-pyruvate allows non-invasive in vivo imaging of TERT expression in brain tumors
    Georgios Batsios1, Celine Taglang1, Meryssa Tran1, Anne Marie Gillespie1, Joseph Costello2, Sabrina Ronen1, and Pavithra Viswanath1
    1Radiology and Biomedical Imaging, UCSF, San Francisco, CA, United States, 2Neurological Surgery, UCSF, San Francisco, CA, United States
    Telomerase reverse transcriptase (TERT) is essential for glioma proliferation and is an attractive therapeutic target. Here, we show that TERT expression in gliomas is linked to higher NADH, an effect that can be non-invasively monitored by deuterium metabolic imaging using [U-2H]pyruvate.
    Lactate production from [U-2H]pyruvate is localized to the tumor region in vivo. (A) Anatomical T2-weighted MRI of a mouse bearing an orthotopic BT88 tumor xenograft. Representative 2H-MR spectra from contralateral normal brain (B) and tumor (C) voxels at the first time point after injection of [U-2H]pyruvate in a mouse bearing an orthotopic BT88 tumor. Metabolic heatmap of the SNR of lactate (D) and the ratio of lactate to post-injection HDO (E) in mouse bearing an orthotopic BT88 tumor. The tumor is delineated by white line.
    Lactate production from [U-2H]pyruvate is higher in orthotopic glioma-bearing mice in vivo. Lactate signal normalized to the ratio of post-injection HDO at each time point to pre-injection HDO is higher in mice bearing orthotopic glioma xenografts relative to tumor-free controls. ** refers to p<0.01, *** refers to p<0.001.
  • Early noninvasive metabolic biomarkers of mutant IDH inhibition in low-grade glioma models
    Marina Radoul1, Donghyun Hong1, Anne Marie Gillespie1, Chloé Najac1, Pavithra Viswanath1, Russell O. Pieper2,3, Joseph Costello2, H. Artee Luchman4, and Sabrina M. Ronen1,3
    1Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, United States, 2Neurological Surgery UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, United States, 3Brain Tumor Research Center, University of California San Francisco, San Francisco, CA, United States, 4Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
    Mutant IDH inhibition monitored in patient-derived glioma using in vivo 1H MRS revealed a decrease in 2HG and increase in Glu and GLX that were associated with subsequent slowdown of tumor growth and survival. This identifies early metabolic biomarkers of mutant IDH inhibition in glioma.
    Figure 4: Representative axial T2-weighted images of control BT257 (A) and SF10417 (B) LGG-bearing mice and corresponding in vivo 1H MRS spectra (black) acquired from 2x2x2mm3 voxel marked in blue as well as the LCModel fit used to quantify the spectra (red) (A, B). Quantification of 2HG, Glu and GLX in control, AG-881- and BAY-1436032-treated tumor voxel at D7 and D15 in BT257 (C) and SF10417 (D) LGG-bearing mice.
    Figure 2: Representative axial T2-weighted images of control, AG-881 and BAY-1436032-treated BT257 (A) and SF10417 (B) LGG-bearing mice at D0, D7 and D15. Temporal evolution of average tumor volume shown as a percentage of D0 in BT257 (C) and SF10417 (D) LGG models.
  • Imaging response to radio-chemotherapy in brain tumor models using [2,3-2H2]fumarate and deuterium magnetic resonance spectroscopic imaging
    Friederike Hesse1, Alan Wright1, Vencel Somai1,2, Flaviu Bulat1,3, and Kevin Brindle1,4
    1Cancer Research UK Cambridge Institute, Cambridge, United Kingdom, 2Department of Radiology, University of Cambridge, Cambridge, United Kingdom, 3Department of Chemistry, University of Cambridge, Cambridge, United Kingdom, 4Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
    Tumor malate production from [2,3-2H2] fumarate increased significantly within seven days of targeted radio-chemotherapy, demonstrating the potential of 2H-labeled fumarate for assessing GB tumor cell death and the early responses of brain tumors to treatment. 
    Figure 1 2H MR spectroscopic measurements of labeled fumarate, malate and water concentrations in A11 (A, B, E,F) and U87 (C,D,G,H) tumors. Tumor spectra were acquired before and 7 days after targeted radio-chemotherapy (10 Gy in total, temozolamide 100 mg/kg). (E – H) Sum of 12 2H spectra recorded over 60 minutes. The [2,3-2H2]fumarate injection (1 g/kg) started 5 min after the start of acquisition of the first spectrum. The peaks were fitted individually using prior knowledge.
    Figure 2 Metabolite concentration maps derived from dynamic 3D CSI images summed over 60 min of signal acquisition following fumarate injection into A11 tumor-bearing mice. The color code represents concentration in mM derived from the ratios of the peak intensities in the malate and fumarate maps to peak intensities in an initial HDO map and corrected for the number of 2H labels per molecule and signal saturation. (A,D) T2-weighted axial slices from a reference 1H image. Concentration maps of (B) fumarate and (C) malate pre-treatment; (E) fumarate and (F) malate 7 days post-treatment.
Back to Top
Digital Poster Session - Cancer: Preclinical & Clinical
Cancer/Spectroscopy/Molecular Imaging/Pre-Clinical
Monday, 17 May 2021 13:00 - 14:00
  • Hyaluronan depletion improved intratumor pO2 and sensitized tumor to radiation therapy in pancreatic cancer model mouse.
    Yu Saida1, Tomohiro Seki2, Shun Kishimoto1, Yasunori Otowa1, Kota Yamashita1, Kazutoshi Yamamoto1, Nallathamby Devasahayam1, Jeffrey R. Brender1, and Murali C. Krishna1
    1Radiation Biology Branch, National Cancer Institute, Bethesda, MD, United States, 2Laboratory of Pharmaceutics, Faculty of Pharmacy and Pharmaceutical Sciences, Josai University, Saitama, Japan
    Multimodal-molecular imaging showed improved oxygenation, increased blood volume, and decreased lactate-to-pyruvate ratio in tumors in response to PEGPH20 treatment. PEGPH20 could enhance the effect of radiotherapy.

    Fig. 3 HP [1-13C] Pyruvate MRI showed decreased Lac/Pyr ratio after PEGPH20 treatment.

    HP [1-13C] Pyruvate MRI was performed in BxPC3-HAS3 tumor bearing mice treated with control buffer or PEGPH20 before and after treatment. A, Representative kinetics of [1-13C] pyruvate and [1-13C] lactate and its anatomical 1H image. B, Lac/Pyr ratio of each tumor are shown by treatment group. Individual values are shown. C, Lac/Pyr ratio change from the baseline (pre-treatment) of each group.

    Fig. 1 EPR imaging showed increased pO2 after PEGPH20 treatment.

    EPR imaging was performed in BxPC3-HAS3 tumor bearing mice treated with control buffer or PEGPH20 before and after treatment. A, Representative oxygen map obtained by EPR imaging and T2-weighted anatomical image. B, Histograms of pO2 distribution within the tumor of each group. C, The mean pO2 of each tumor shown by treatment group. D, pO2 change (%) from the baseline (pre-treatment) of each group.

  • Multimodal molecular imaging assessment of changes in tumor microenvironment in response to combination of Evofosfamide and GEM
    Yasunori Otowa1, Kota Yamashita1, Yu Saida1, Kazutoshi Yamamoto1, Jeffery R Brender1, Nallathamby Devasahayam1, Murali C. Krishna1, and Shun Kishimoto1
    1National Cancer Institute, Bethesda, MD, United States
    still in progress
    Permeability of Gd-DPTA into SU.86.86 and MIA Paca-2 tumors before treatment on Day 1 and 1 hour after treatment on Day 5 evaluated by DCE-MRI. (A) (B) (D) (E)Kinetics of the Gd-DTPA incorporation into SU.86.86 or MIA Paca-2 tumors treated with combination of evofosfamide and GEM or vehicle. (C) Comparison of Ktrans in combination of evofosfamide and GEM and control treated tumor in SU86.86 tumor (*P=0.041). (F) Comparison of Ktrans in combination of evofosfamide and GEM and control treated tumor in MIA Paca-2 tumor (***P<0.001).
    Blood volume (%) changes calculated using MRI with the blood pooling T2 contrast agent USPIO. (A) (B) (C) (D) The images of blood volume on Day 1 and Day 5 in SU.86.86 tumors. (E) Comparison of blood volume on Day 5 relative to Day1 in SU.86.86 tumors (**P=0.004). (F) (G) (H) (I) The images of blood volume on Day 1 and Day 5 in MIA Paca-2 tumors. (E) Comparison of blood volume on Day 5 relative to Day1 in MIA Paca-2 tumors (*P=0.036).
  • Iron oxide-based Enzyme Mimic Nanocomposite for Dual-Modality Imaging Guided Chem-phototherapy and Anti-tumor Immunity Against Breast Cancer
    Xiuhong Guan1, Jiali Cai2, Xiangyu Xiong1, Hong Liu2, Shihui Huang2, Sheng Wang2, Chuanqi Sun1, Yi Sun3, Tianjing Zhang4, Guoxi Xie1, and Zhiyong Wang2
    1Department of Biomedical Engineering, Guangzhou Medical University, Guangzhou, China, 2School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, China, 3Siemens Healthineers, Shanghai, China, 4Philips Healthcare, Guangzhou, China
    This study reports a kind of functional nanocomposite, superparamagnetic iron oxide nanocrystals (SPIO@NC), exhibiting NIR-II and MR dual-modal imaging guided chem-phototherapy and anti-tumor immunity against triple-negative breast cancer.
    Schematic illustration of experimental flow. Schematic illustration of SPIO@NC synthesis and experiments in mice in vitro and in vivo.
    Fig. 4 (A)(B)Representative flow cytometric analysis displaying the absolute percentage of (A) M1 (MHC-II+) and M2 (CD206+) macrophages in CD45+CD11b+ tumor cells, and (B) CD8+ T cells and CD4+ T cells in CD45+CD3+ tumor cells from mice with different treatments at 7 days. (n = 5). (C) Representative immunofluorescence imaging of Treg cells (CD4+FoxP3+) in tumor from mice received various treatments on 7th days. Scale bar, 30μm. (D)(E) Real pictures of tumors(D) and (E) after dissection from mice received different treatments.
  • Precision MRI (pMRI) of Liver Metastasis enabled by Protein MRI contrast agents
    Jenny Yang1, Mani Salarian2, Hua Yang3, Shanshan Tan4, Oluwatosin Y Ibhagui4, Jingjuan Qiao4, Zongxiang Gui4, and Hans E Grossniklaus3
    1Chemistry, Georgia State University, Atlatna, GA, United States, 2Chemistry, Georgia State University, Atlanta, GA, United States, 3Emory University, Atlanta, GA, United States, 4Georgia State University, Atlanta, GA, United States
    We anticipate that pMRI will demonstrate significantly-improved imaging sensitivity and accuracy and will enable detection of liver metastasis at a much earlier stage, potentially leading to improved treatment responses.
    Figure 4. . MR images of metastatic mice models with ProCA32.CXCR4 administration. A. Comparison of MRI images of metastatic mice models including M20-09-196, OMM2.3, and OCM1 before and after administration of ProCA32.CXCR4. B. Zoom-in view of the metastases from M20-09-196, OMM2.3, and OCM1 mouse models; MRI signal-noise-ratio (SNR) of metastases following the ProCA32.CXCR4 administration.
    Figure 2. A Detection of UM liver metastasis using ProCA32.collagen (top), non-targted ProCA32 (middle) and Eovist (bottom) at 7T. Detected liver metastasis are verified by H&E and Sirius red staining (B) and UM markers HMB45 and S100 staining (C). The tumor liver contrast to noise ratio is further increased by inversion recovery pulse sequence showed in the figure on right.
  • Imaging ascorbate-mediated oxidative stress in PDX models of pancreatic cancer
    Nathaniel Kim1, Arsen Mamakhantan1, Kristin Granlund1, Elisa de Stanchina2, Manish Shah3, Lewis Cantley4, and Kayvan R. Keshari1
    1Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, United States, 2Antitumor Assessment Core Facility, Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States, 3Weill Cornell Medicine, New York-Presbyterian Hospital, New York, NY, United States, 4Meyer Cancer Center, Department of Medicine, Well Cornell Medical College, New York, NY, United States
    Hyperpolarized dehydroascorbic acid was used to measure changes in redox in KRAS and BRCA mutant PDX models of pancreatic cancer. Increasing the hyperpolarized lifetime via D2O solvation in awake mouse injections allowed for changes in oxidative stress to be measured upon ascorbate therapy.
    Figure 3. Representative HP 13C MRS of (A) vehicle and (B) ascorbate-treated KRAS PDX model of pancreatic cancer. Metabolic conversion of DHA to ascorbate in pancreatic cancer PDXs changes after 1 week treatment of twice daily ascorbate.
    Figure 2. Tumor growth curves for patient derived xenograft models of KRAS and BRCA driven cancer. After 2 weeks of tumor implantation, mice were randomly divided into two groups. One group was treated with freshly prepared vitamin C in 400 μL of PBS (4 g/kg) twice a day via IP injection (KRAS, n= 7; BRCA, n = 7). Control group mice were treated with PBS using the same twice a day dose (KRAS, n = 7; BRCA, n = 7). Tumor growth change for the KRAS PDX tumors is significant after 21 days and significant after 4 days for the BRCA PDX tumors.
  • 3D Free-breathing Multitasking T1-T2 Mapping in Small Animals on a 3-Tesla System: A Preliminary Study on a Murine Model with Liver Metastasis
    Nan Wang1, Jingjuan Qiao2, Zhijun Wang3, Pei Han1,4, Hsu-Lei Lee1, Sen Ma1, Hui Han1, Zhaoyang Fan1, Anthony G. Christodoulou1, Ekihiro Seki3, Stephen Pandol5, Debiao Li1, Jenny Yang2, and Yibin Xie1
    1Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States, 2Chemistry Department, Georgia State University, Atlanta, GA, United States, 3Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States, 4Bioengineering, University of California, Los Angeles, Los Angeles, CA, United States, 5Division of Digestive and Liver Diseases, Cedars-Sinai Medical Center, Los Angeles, CA, United States
    A Multitasking technique was proposed for mouse abdominal imaging at 3T, achieving 3D motion-resolved acquisition and simultaneous T1 T2 mapping within 10 minutes. It produced better images with vastly reduced scan time on a murine model with liver metastasis of colorectal cancer.
    Figure 4: T1-T2-weighted images and T1 T2 maps from conventional methods and from Multitasking acquired on an HFD mouse at week 2 with ProCA.collagen1 injection. Multitasking consistently showed improved image quality. The tumor were labeled by red dashed boundary on T2W TSE. Tumor showed higher T1 and T2 values compared to normal liver parenchymal, which was consistent with conventional methods and Multitasking. Some breathing artifacts on conventional images were labeled by white arrows
    Figure 3: T1-T2-weighted images and T1 T2 maps from conventional methods and from Multitasking acquired on an LFD mouse at week 1 with Eovist injection. In each imaging session, conventional image series took about 45 to 50 minutes in total, while Multitasking can generate T1-T2-weighted images and T1 T2 maps in one single 10-min scan. Moreover, Multitasking produced improved image quality in both weighted images and quantitative maps. Some breathing artifacts on conventional images were labeled by white arrows
  • Deuterium metabolic imaging (DMI) of glucose highlights pancreatic cancers in two mice models
    Stefan Markovic1, Tangi Roussel2, Keren Sasson3, Dina Preise3, Lilach Agemi3, Avigdor Scherz3, and Lucio Frydman1
    1Department of Chemical and Biological Physics, Weizmann Institute of Science, Rehovot, Israel, 2Center for Magnetic Resonance in Biology and Medicine, Marseille, France, 3The Moross Integrated Cancer Research Center, Weizmann Institute of Science, Rehovot, Israel
    Deuterium Metabolic Imaging was used to follow the metabolic conversion of 2H6,6’-glucose in a pancreatic cancer mouse model at 15.2T. Lactate was produced exclusively in the tumors, leading to the marking of the latter within the abdomen.
    DMI data collected at the indicated stages following the intravenous administration of 2H6,6’-glucose to a pancreatic cancer mouse. Metabolic maps of 2H6,6’-glucose (A) and its metabolic products 2H3,3’-lactate (B) and 2H-water (C) are here shown as absolute concentration colormaps. The anatomical 1H image on top of which all 2H data are shown is depicted in (D). Concentrations for glucose and water (E) and for the lactate (F) are shown for the entire time series.
    2H NMR spectra arising at the indicated times after an intravenous 2H6,6’-glucose administration into a pancreatic cancer mouse model. (B) and (C) show organ-specific 2H spectra extracted from the CSI data at the indicated sites. Signals for 2H6,6’-glucose and its metabolic products water and lactate are indicated by letters G, W and L respectively.
  • In-Vivo Cell Tracking of Murine Natural Killer Cells in Lymphoma by Fluorine-19 MRI
    Lawrence Lechuga1, Sean B Fain1,2,3,4, Christian M Capitini3,4,5, and Matthew H Forsberg5
    1Medical Physics, University of Wisconsin, Madison, Madison, WI, United States, 2Radiology, University of Wisconsin, Madison, Madison, WI, United States, 3Biomedical Engineering, University of Wisconsin, Madison, Madison, WI, United States, 4Carbone Cancer Center, University of Wisconsin, Madison, Madison, WI, United States, 5Pediatrics, University of Wisconsin, Madison, Madison, WI, United States
    PFPE labeled NK cells were detected and quantified in 3 lymphoma bearing mice out to 6 days post injection by 19F MRI. Quantification indicates that 87% and 70% were detectible at days 0 and 6. Postmortem flow cytometry verified that NK cells maintain label and viability out to 6 days post injection.
    Representative mouse composite magnitude images on Day 0, 3, and 6 after intratumoral injection of 5.3 x 105 NK cells into syngeneic EL4 lymphomas. Images were scaled against their own noise to place into units of pixel SNR. The 19F signal mitigating from labeled cells is detectible within the tumor in all three images. White arrow indicates the PFPE reference vial of known spin density.
    In vivo NK cell quantification results indicate the calculated number of PFPE-red labeled GFP+ NK cells within the tumor volume of each mouse. Difference in time points are not statistically significant.
  • Comparison of Tumour pH Environment and Glycolysis Measurements in a C6 Rat Model of Glioma
    Qi Qi1,2,3, Matthew Fox1,4, Robert Bartha2,5,6, Miranda Bellyou5, Lise Desjardins7, Lisa Hoffman1,2,8, Alex Li5, Andrew McClennan1,2, Ting Yim Lee1,2,3,5,6, and Jonathan D Thiessen1,2,3,6
    1Lawson Imaging, Lawson Health Research Institute, London, ON, Canada, 2Medical Biophysics, Western University, London, ON, Canada, 3Molecular Imaging, Western University, London, ON, Canada, 4Physics and Astronomy, Western University, London, ON, Canada, 5Robart Research Institute, London, ON, Canada, 6Medical Imaging, Western University, London, ON, Canada, 7Lawson Health Research Institute, London, ON, Canada, 8Anatomy and Cell Biology, Western University, London, ON, Canada
    This study demonstrates the capability of simultaneous measurements of pHi and pHe using CEST MRI, a more complete picture of the tumour pH environment, and explores the intrinsic relationship between tumour glycolysis and its pH environment.
    Figure 2: Illustrative examples of baseline (a) and post-injection (b) AACID maps; baseline (c) and post-injection (d) acidoCEST maps. All maps are overlaid on top of the corresponding T2-weighted MRI. The tumour region is delineated with the red-dotted line, and the peri-tumour region is contoured with the light-blue-dotted line using the T2-weighted MRI.
    Figure 1: An illustrative example of the FDG-PET image of the last frame (SUV, a), time-activity curve from the tumour region (b) delineated with red-dotted line, and the arterial input function (AIF) from the left ventricle of the heart delineated with white-dotted line (c, d). The axial SUV map is overlaid on top of the corresponding T2-weighted MRI.
  • MRS based biomarkers of IDH1 mutant glioma response to the BAY-1436032 IDH inhibitor
    Donghyun Hong1, Georgios Batsios1, Pavithra Viswanath1, Anne Marie Gillespie1, Russell O Pieper2,3, Joseph Costello2, and Sabrina M Ronen 1,3
    1Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, United States, 2Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, United States, 3Brain Tumor Research Center, University of California San Francisco, San Francisco, CA, United States
    Here we treated mutant IDH1-expressing cells with the emerging inhibitor BAY-1436032 and identified translatable MRS based metabolic biomarkers of mutant IDH1 inhibition using 1H and hyperpolarized 13C spectroscopy.
    Figure 1. Representative 1H MRS spectra of control and treated NHAIDH1mut cells.
    Figure 3. (A) shows fluxed from the hyperpolarized [1-13C]α-ketoglutarate to glutamate between control (blue) and the BAY-1436032 treated (orange) cells. A significant increase in glutamate was observed in the BAY-1436032 cells (B)
  • 31P MRSI in tumor-bearing mice at 9.4T
    Vanessa L. Franke1, Justyna Platek1, Philip S. Boyd1, Stephanie Laier2, Karin Mueller-Decker2, Andrey Glinka3, Mark E. Ladd1, Steffen Goerke1, Peter Bachert1, and Andreas Korzowski1
    1Division of Medical Physics in Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany, 2Center for Preclinical Research, Core Facility Tumor Models, German Cancer Research Center (DKFZ), Heidelberg, Germany, 3Division of Molecular Embryology, German Cancer Research Center (DKFZ), Heidelberg, Germany
    31P MRSI with large spatial coverage in mice at B0=9.4T with a spatial resolution of (2.5x2.5x7.5)mm³ obtained in 50 minutes is feasible and enables the quantification of signals in diseased tissue, while maintaining the separation to healthy tissue
    Figure 4: Summed 31P spectra from the muscle (left) and tumor ROI (right) with the corresponding fitted signal (red line) acquired with protocol 2 (Fig.3). Acquisition parameters: TR=300ms, α=45°, Δf=6060 Hz, 1024 time points, postprocessing with a 40-Hz Gaussian filter in time domain. The following resonances were resolved: PCr, ATP, Pi, PC, and Phosphoethanolamine (PE). Note the different scales of the y-axis in both spectra.
    Figure 3: Transversal map of the fitted PCr amplitude overlaid on the morphological 1H image in the slice covering the tumor. Data was acquired with protocol 2: matrix size=10x10, FOV=(25x25)mm², Hamming-weighted k-space averaging with 400 central averages, postprocessing with zerofilling-factor 2. The tumor ROI (blue) was drawn on the marginal part of the tumor to reduce signal contamination from muscle. The muscle ROI (green) includes the same number of voxels as the tumor ROI.
  • MR elastography reveals a marked increase in breast cancer viscoelasticity in vivo following hyaluronan degradation by PEGPH20
    Emma L. Reeves1, Jin Li1,2, Konstantinos Zormpas-Petridis1, Jessica K. R. Boult1, James Sullivan1,3, Craig Cummings1, Barbara Blouw4, David Kang4, Ralph Sinkus5, Yann Jamin1, Jeffrey C. Bamber1, and Simon P. Robinson1
    1Radiotherapy & Imaging, Institute of Cancer Research, London, United Kingdom, 2Institutes of Brain Science, Fudan University, Shanghai, China, 3Royal Marsden NHS Foundation Trust, Sutton, United Kingdom, 4Halozyme Therapeutics, San Diego, CA, United States, 5Division of Imaging Sciences and Biomedical Engineering, King's Health Partners, St Thomas's Hospital, London, United Kingdom
    MRE revealed a ~80% increase in MDA-MB-231 LM2-4 tumour viscoelasticity following hyaluronan (HA) degradation by PEGPH20. However, no PEGPH20-induced change in viscoelasticity occurred in 4T1 or 4T1/HAS3 tumours. Hence, MRE is unlikely to provide a robust biomarker of HA degradation.
    Figure 1. Anatomical T2-weighted (T2w) MRI and parametric maps of Gd, Gl, |G*| and Y for representative 4T1, 4T1/HAS3 and MDA-MB-231 LM2-4 tumours prior to and 24 hours after treatment with PEGPH20 (1 mg/kg). The tumour is delineated by a white dashed line.
    Figure 3. Anatomical T2-weighted (T2w) MRI and aligned tissue sections stained with H&E, HTI-601 (hyaluronan/HA) and picrosirius red (collagen I & III). Representative saline and PEGPH20 treated tumours are shown for each tumour model (4T1, 4T1/HAS3 and MDA-MB-231 LM2-4). The PEGPH20 treated tumours are the same as those shown in Figure 1. The whole section images highlight the close matching of the histology with MRI. Representative high-power images (20x) are inset next to each whole tumour section.
  • Metabolomic Characterization of renal cell carcinoma patient-derived xenografts and derived Tissue Slice Cultures
    Deepti Upadhyay1, Jinny Sun1, Joao Piraquive Agudelo1, Hongjuan Zhao2, Rosalie Nolley2, Robert Bok1, James D. Brooks2, Donna M. Peehl1, John Kurhanewicz1, and Renuka Sriram1
    1Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, United States, 2Department of Urology, Stanford University, Stanford, CA, United States
    Metabolic characterization of renal cell carcinoma patient derived-xenografts and tissue slice cultures using stable isotope resolved metabolism showed metabolic heterogeneity among PDXs and TSCs of different clinical and pathological stages.
    Figure 4. 13C Isotopomer modeling of [U-13C] glucose-labeled of PDX047 and TSC047 using tcaCALC software. (PDH - Pyruvate dehydrogenase, PK-Pyruvate Kinase, Y-denotes anaplerotic denotes, Ypc - Pyruvate Carboxylase and Ys- anaplerosis via succinyl CoA, glutaminolysis)
    Figure 3: Comparison of (A) steady state concentration and (B) Fractional enrichment of glutamate among PDX model, while (C) & (D) represent steady state concentration and Fractional enrichment of glutamate, respectively among TSCs model. *p<0.05, **p<0.005, ***p< 0.0005, n=3 for all groups except TSC072 (n=2)
  • Glioma Genetic Diagnosis Software for Detection of IDH and TERTp Mutations based on 1H MR Spectroscopy and Mass Spectrometry
    Abdullah Bas1, Banu Sacli-Bilmez1, Gokce Hale Hatay1, Alpay Ozcan2,3, Cansu Levi4, Ayca Ersen Danyeli3,5, Ozge Can3,6, Cengiz Yakicier3,7, M.Necmettin Pamir3,8, Koray Ozduman3,8, Alp Dincer3,9, and Esin Ozturk-Isik1,3
    1Institute of Biomedical Engineering, Bogazici University, İstanbul, Turkey, 2Electrical and Electronics Engineering, Bogazici University, Istanbul, Turkey, 3Center for Neuroradiological Applications and Reseach, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey, 4Department of Medical Biochemistry, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey, 5Department of Medical Pathology, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey, 6Department of Medical Engineering, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey, 7Department of Molecular Biology and Genetics, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey, 8Department of Neurosurgery, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey, 9Department of Radiology, Acıbadem Mehmet Ali Aydinlar University, Istanbul, Turkey
    Glioma Genetic Diagnosis Software was developed for noninvasive detection of IDH and TERTp mutations in gliomas. The machine-learning models in the tool were trained with proton magnetic resonance spectroscopy and mass spectrometry data of 237 gliomas.
    Figure 3: Visualization of the data.
    Table 1: Performance metrics of the default models in the modules in Normal User part (*Due to class imbalance, synthetic data was generated with ADASYN)
  • Tumour vascular response to the FGFR inhibitor derazantinib assessed using susceptibility-contrast MRI with ferumoxytol
    Jessica K.R. Boult1, Mahmoud El Shemerly2, Felix Bachmann2, Laurenz Kellenberger2, Heidi Lane2, Paul McSheehy2, and Simon P. Robinson1
    1The Institute of Cancer Research, Sutton, United Kingdom, 2Basilea Pharmaceutica International Ltd, Basel 4005, Switzerland
    FGFR inhibitor derazantinib causes dose-dependent inhibition of endothelial cell proliferation, VEGFR signalling and vascular permeability in mouse skin, and induces a reduction in fractional blood volume, as assessed by susceptibility-contrast MRI, in colorectal tumour xenografts.
    Figure 3. T2w images and paramagnetic maps of baseline R2*, USPIO-induced ΔR2* and fractional blood volume (fBV) from representative athymic nude mice pre and post 48h treatment with vehicle, 80mg/kg DZB daily or 50mg/kg vatalanib twice daily.
    Figure 4. Quantification of baseline R2* and fractional blood volume (fBV) in all mice pre and post 48h treatment with vehicle, 80mg/kg DZB daily or 50mg/kg vatalanib twice daily. DZB and vatalanib induced significant reductions in fBV (p<0.05, 2-way repeated measures ANOVA).
  • Evaluating the utility of DCE-MRI in differentiating brain tumours using the extended Tofts and the Shutter Speed Model
    Sourav Bhaduri1, Samantha Mills2, Mark Radon2, Michael Jenkinson3, and Harish Poptani1
    1Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom, 2Department of Neuroradiology, The Walton Centre NHS Foundation Trust, Liverpool, United Kingdom, 3Department of Neurosurgery, The Walton Centre NHS Foundation Trust, Liverpool, United Kingdom
    We demonstrate the potential utility of DCE-MRI derived pharmacokinetic parameters in differentiating brain tumour types using the extended Tofts and the Shutter Speed Model.
    Fig 1: Model fitting using A. extended Tofts and B. SSM in PCNSL. The same is shown for GBM in C and D. The original AIF curve is shown in yellow and its biexponential fit shown in black. The tissue uptake is shown in blue with its fitting shown in red.

    Fig 2: Ktrans maps from the extended Tofts model from a patient with PCNSL (A), Gr III Glioma (B), GBM (C) and Metastasis (D). Box plots demonstrating the Ktrans (min-1, E), ve (F) and vp (G) values using extended Tofts model demonstrate highest values in metastasis, while PCNSL patients demonstrate lower values.

  • Estimation of capillary level input function for abbreviated breast Dynamic Contrast-Enhanced MRI using deep learning approach
    Jonghyun Bae1,2,3, Zhengnan Huang1,2,3, Florian Knoll2,3, Krzysztof Geras2,3, Terlika Sood2,3, Laura Heacock2,3, Linda Moy2,3, Li Feng4, and Sungheon Gene Kim5
    1NYU School of Medicine, New York, NY, United States, 2Center for Advanced Imaging Innovation and Research, New York, NY, United States, 3Center for Biomedical Imaging, NYU, New York, NY, United States, 4Icahn School of Medicine at Mount Sinai, New York, NY, United States, 5Weill Cornell Medicine, New York, NY, United States
    Our proposed AI-based approach to estimate CIF has demonstrated its accuracy in estimating Pharmacokinetic parameters to aid the diagnosis of the breast cancer in the clinical setting, while eliminating the need for any manual selection of AIF.
    (a) A patch of DCE data has been rearranged to 2-dimensional matrix X = (n t), where n is the number of voxel (9 in our design) and t is the number of temporal frames. (b) Schematic diagram for the deep learning network. The total of twelve 2D-convolutional layers were connected in series and each convolutional layer is linked with Rectified Linear Unit activation function. After two layers, a skipping connection was made and filters were concatenated together, just like in the Residual Network
    Receiver Operating Characteristic (ROC) curve for the estimated Fp from the PKM analysis of the clinical data. Fp estimation from 3 approaches using (1)the case-specific AIF(Ca), (2)the population-averaged AIF, and (3)the predicted Cp with the population-averaged AIF(Cp+Ca,pop) was used to assess the diagnostic performance in predicting the malignancy of breast cancer. Fp estimation from our proposed model yielded the highest AUC.
  • Commonality and complexity of systemic metabolic dysregulation caused by cancer and cancer-induced cachexia
    Santosh Kumar Bharti1, Raj Kumar Sharma1, Paul T Winnard1, Marie-France Penet1, and Zaver M. Bhujwalla1,2,3
    1Div. of Cancer Imaging Research, The Russell H. Morgan Dept of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States, 2Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, United States, 3Radiation Oncology and Molecular Radiation Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
    Identification of systemic metabolic dysregulation caused by cancer and cancer-induced cachexia in PDAC cachexia mice model using high-resolution 1H MRS
    Figure 1: Representative 1H MR spectra acquired from aqueous phase tissue extracts of spleen obtained from normal mice and cachectic (Pa04C ) and non-cachectic (Panc1) tumor bearing mice.
    Figure 2: Lung, liver, heart, kidney and spleen weights in control mice (black) as compared with Panc1 (green) and Pa04C (red) tumor‐bearing mice. In all panels, means of cohorts are shown as rectangles bar. P-value less that ≤ 0.05 are considered statistically significant.
  • Wavelet Oversampling for Imbalance Childhood Brain Tumour Classification
    Dadi Zhao1,2, James T. Grist1,2, Heather E.L. Rose1,2, Yu Sun1,2, and Andrew C. Peet1,2
    1Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom, 2Department of Oncology, Birmingham Children's Hospital, Birmingham, United Kingdom
    Wavelet oversampling showed significantly improved classification performance of childhood brain tumours through metabolite profiles from 1H-MRS.
    Illustration showing the procedure of generating oversampled proton magnetic resonance spectroscopy (1H-MRS) from the raw 1H-MRS by using Wavelet OverSampling (WvOS). Abbreviations: SNR, signal-to-noise ratios; ψ, wavelet basis.
    Boxplots showing the balanced classification accuracy for the 1.5T cohort of childhood brain tumours derived through linear discriminant analysis (A, C) or supper vector machine (B, D) with leave-one-out (A-B) or six-fold (C-D) cross validation.
  • Tumorous Tissue Characterization in Diffuse Glioma Based on 1H-MRS Data Employing 1D Convolutional Neural Networks
    Farzad Alizadeh1,2, Anahita Fathi Kazerooni3,4, Hanieh Bahrampour5, Hanieh Mobarak Salari1,2, and Hamidreza Saligheh Rad1,2
    1Department of Medical Physics and Biomedical Engineering, Tehran university of Medical Science, Tehran, Iran (Islamic Republic of), 2Quantitative MR Imaging and Spectroscopy Group, Research Center for Molecular and Cellular Imaging, Tehran, Iran (Islamic Republic of), 3Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, United States, 4Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States, 5Biomaterials Engineering, School of Metallurgy and Materials Engineering, Iran University of Science and Technology, Tehran, Iran (Islamic Republic of)
    Convolutional neural networks are able to differentiate brain tumorous tissue subregions based on 1H-MRS data with acceptable accuracy scores.
    Our proposed Resnet-based convolutional neural network architecture applied to classify five tissue types in diffuse glioma.
    T1-w image of a patient with a diffuse glioma (a). Chemical Shift Image (CSI) of the patient (b). Rotated overlaid image of CSI grid on T1-w image (c). color coded grid of CSI in order to exact localization of the signals (d).
Back to Top
Digital Poster Session - Cancer: Clinical
Cancer/Spectroscopy/Molecular Imaging/Pre-Clinical
Monday, 17 May 2021 13:00 - 14:00
  • Radiomics Based Classification of Ependymoma and High Grade Glioma Using Multimodal MRI
    Apoorva Safai1, Sumeet Shinde1, Manali Jadhav1, Tanay Chougule1, Abhilasha Indoria2, Manoj Kumar2, Vani Santosh2, Shumyla Jabeen2, Manish Beniwal2, Subhash Konar2, Jitender Saini2, and Madhura Ingalhalikar1
    1Symbiosis International University, Pune, India, 2National Institute of Mental Health and Neurosciences, Bangalore, India
    Quantitative radiomic markers such as texture and first order statistics from multimodal MRI can capture intricate and complementary information and thus aid in a robust multiclass tumor classification of STEE and HGG subtypes.
    Fig-2:Processing pipeline implemented for radiomics analysis and classification of tumor subgroups
    Fig-5: Feature importances obtained using SVM coefficient scores on multimodal feature set
  • Creating a radiomic signature for H3K27M mutation in midline glioma on multimodal MRI
    Manali Balasaheb Jadhav1, Richa Singh Chauhan2, Priyanka Tupe Waghmare3, Archit Rajan4, Abhilasha Indoria2, Jitender Saini5, Vani Santosh6, Madhura Ingalhalikar4, and Subhas Konar7
    1Symbiosis Center for Medical Image Analysis, Pune, India, 2Radiology, National Institute of Mental Health and Neuroscieces, Bengaluru, India, 3Symbiosis Institute on Technology, Pune, India, 4Symbiosis Centre for Medical Image Analysis, Pune, India, 5Radiology, National Institute of Mental Health and Neuroscieces, Pune, India, 6Neuropathology, National Institute of Mental Health and Neuroscieces, Bengaluru, India, 7Neurosurgery, National Institute of Mental Health and Neuroscieces, Bengaluru, India
    Our random forest classifier with radiomics computed from multi-modal MRI provides high discriminative accuracy in predicting H3K27M mutation in midline glioma.
    Figure 1: Processing pipeline for radiomics analysis and classification for histone H3K27M mutation
    Figure 4: Box plot for top 10 most important features obtained using random forest classifier
  • Using Variable Flip Angle (VFA) and Modified Look Locker Inversion Recovery (MOLLI) T1 Mapping in Clinical OE-MRI
    Emma Bluemke1, Ambre Bertrand1, Kwun-Ye Chu2,3, Nigar Syed2, Andrew Murchison3,4, Tessa Greenhalgh3,5, Brian Burns6, Martin Craig7, Nia Taylor3, Ketan Shah2,3, Fergus Gleeson2,3, and Daniel Bulte1
    1University of Oxford, Oxford, United Kingdom, 2MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom, 3Radiotherapy Department, Oxford University Hospitals NHS, Oxford, United Kingdom, 4Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom, 5University Hospital Southampton NHS FT, Southhampton, United Kingdom, 6GE Healthcare, Menlo Park, CA, United States, 7University of Nottingham, Nottingham, United Kingdom
    VFA T1 was higher than the MOLLI T1 by 27%. No significant difference in standard deviation within tumour ROIs from VFA vs MOLLI. VFA was a more robust method, considering patient motion in the z-plane rendered MOLLI unusable for OE-MRI analysis.
    Example VFA and MOLLI T1 maps for 6 subjects (displaying a comparable slice). The field-of-view of the images has been cropped for ease of display.
    (A-B) The mean T1 in the tumour ROI of all subjects. The error bars represent the standard deviation of T1 values within the tumour ROI. (C) The mean VFA T1, normalized by the respective mean MOLLI T1 for each subject to illustrate the relative change in T1 clearly.
  • Assessment the Preponderant Diagnostic Performances of Oligometastatic Prostate Cancer Using DCE-MRI of Tofts Model
    SHUANG MENG1, Ailian Liu1, Lihua Chen1, Qinhe Zhang1, Qingwei Song1, and Yunsong Liu1
    1The First Affiliated Hospital of Dalian Medical University, Dalian, China
    In this study, we evaluated the diagnostic performances of Tofts model (TM) in assessing oligometastatic prostate cancer (PCa). The results showed that transfer constant (Ktrans) can differentiate the diagnosis oligometastatic and widely metastasis PCa ((OR (95CI)<0.001,95% CI:0.000-0,028), and the AUC (95CI)= 0.77). Ktrans combined with clinical characteristics (such as age and prostate specific antigen (PSA)) had the higher diagnostic efficiency (AUC (95CI)=0.958). Therefore Ktrans has the potential to assess the aggressiveness of PCa . And Ktrans combined age and PSA maybe have the higher diagnostic efficiency for oligometastatic and widely metastasis PCa
    Figure 1 A 60-year-old male with localized lesions PCa.ROI was manually placed on the local lesion of DCE map (A). The Ktrans, Kep and Ve maps of TM were shown(B-D).
    Figure 2.ROC curves show performangces of Ktrans individually and combination of age and PSA to differentiate between oligometastatic and widely metastasis PCa.
  • Evaluating the cytokeratin 19 (CK-19) status via neural network model established by the SWI-derived radiomics features
    Zhijun Geng1, Yunfei Zhang2, Chuanmiao Xie1, and Yongming Dai2
    1Sun Yat-sen University Cancer Center, Guangzhou, China, 2Central Research Institute, United Imaging Healthcare, Shanghai, China
    Deep learning based neural network model established with SWI-derived radiomics features holds great potential in evaluating the prognostic markers of HCC.
    Fig. 3. The structure of ANN model
    Fig. 4 Diagnostic performance of ANN model for identifying the CK-19 status.
  • Improved gastric T2WI imaging by an TSE corrected HASTE sequence: a comparation study with conventional HASTE, TSE and BLADE-TSE sequences on 3T
    Xiaosheng Xu1, Qinglei Shi2, Weishuai Wang3, Jia Wei4, Li Yang4, Qian Xu4, and Gaofeng Shi4
    1The Fourth Hospital of Hebei Medical University, Shijiazhuang, China, 2MR Scientific Marketing,Siemens Healthcare, Beijing, China, 3MR Scientific Marketing,Siemens Healthcare, JINAN, China, 4The Fourth Hospital of Hebei Medical University, shijiazhuang, China
     the HASTE sequence demonstrate good performance in gastric cancer diagnosis with much less motion artifacts and fast acquisition, which may help patients with poor respiratory curve or cannot hold breath

    Fig 1, male, 56 years old with gastric cancer, (A) image acquired by TSE-HASTE with a subjective score of 5 points; (B) image acquired by conventional HASTE sequence sequence with a subjective score of 5 points; (C) image acquired by TSE- BLADE sequence with a subjective score of 3 points; (D) image acquired by conventional TSE sequence with a subjective score of 3 points.

    Fig 2, male, 63 years old with gastric cancer with irregular breathing rhythm, (A) image acquired by TSE-HASTE with a subjective score of 5 points; (B) image acquired by conventional HASTE sequence sequence with a subjective score of 4 points; (C) image acquired by TSE- BLADE sequence with a subjective score of 2 points; (D) image acquired by conventional TSE sequence with a subjective score of 1 points.
  • Deep learning prediction for clear cell renal carcinoma cancer compared with human and radiomics analysis
    Junyu Guo1, Lauren Hinojosa1, Yin Xi1, Keith Husley1, and Ivan Pedrosa1
    1Radiology, UT southwestern medical center, Dallas, TX, United States
    Radiomics and deep learning technique have the potential to facilitate the prediction of clear cell renal carcinoma cancer (ccRCC) subjects even using T2w images only. These results were compared against previously reported performance of the clear cell likelihood score (ccLS) criteria.
    Figure 1. Receiver operating curves (ROC) for three models using radiomics and results of deep learning and human prediction. DL1: deep learning model using a single T2w slice; DL2: deep learning model using a T2w slice and its tumor and kidney masks. ccLS 4/5 and ccLS 3/4/5: clear cell likelihood score 4 and 5, or 3, 4 and 5 from human readers based on multiparametric MRI.
    Table 2
  • The value of preoperative prediction of ki67 and P53 expression in DCE-MRI texture analysis of rectal cancer
    Yuhui Liu1, Ailian Liu1, and Mingxiao Wang1
    1The First Affiliated Hospital of Dalian Medical University, Dalian, China, Dalian, China
     DCE-MRI texture analysis of rectal cancer can  predict  the value of Ki67,P53 expression status before operation.
    Figure 1 A 52-year-old male patient with the rectal tubular adenocarcinoma. T2 image (a), Ktrans image (b), Kep image (c) and Ve image (d) were showed.
    Figure2 Comparison of ROC Curves of the parameters with differences between the Ki67 high/low differentiation,ki67 analyzes the texture analysis of the three post-processing images of Ktrans, Ve, and Kev. After the statistically different parameters are combined, the combined diagnostic performance is obtained,the AUC was 0.923, with the sensitivity of 90.5% and the specificity of 50%.
  • Diagnostic performance of machine learning-based MRI for posterior fossa tumors: a meta-analysis
    Chen Chen1, Fabao Gao1, and Xiaoyue Zhou2
    1Department of Radiology, West China Hospital, Chengdu, China, 2MR Collaboration, Siemens Healthineers Ltd., Shanghai, China
    Machine learning demonstrated excellent diagnostic performance for prediction of PFTs, especially for MB vs non-MB and PA vs non-PA. MRI sequences, algorithms, region of interest, and feature extraction were the main factors affecting the diagnostic performance of machine learning.  
    Table 1 Baseline characteristic of included studies
    Fig. 3 Forest plot of single studies for the pooled DOR and a represents EP vs non-EP, b MB vs non-MB, c PA vs non-PA.
  • Automatic segmentation of glioma based on MRI K-space data
    Yikang Li1,2,3, Zhan li Hu1,2,3, Sen Jia1,2,3, Wenjing Xu4, Zongyang Li1,2,3, Hairong Zheng1,2,3, Xin Liu1,2,3, and Na Zhang1,2,3
    1Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China, 2Key Laboratory for Magnetic Resonance and Multimodality Imaging of Guangdong Province, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China, 3CAS key laboratory of health informatics, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China, 4Faculty of Information Technology, Beijing University of Technology, Beijng, China
    We proposed a new method that omits the Fourier transformations and directly makes segmentations on K space data.
    Table 1. The testing dataset results. Mean Dice, PPV, sensitivity, and Hausdorff are shown in the table. EN is enhancing tumor core. WT is the whole tumor part. And the TC is tumor core.
    Figure 2. A typical segmentation example with ground truth and predicted labels on 2D MRI slices. The whole tumor (WT) part contains all visible labels (all green, red and yellow labels), the tumor core (TC) part contains red and yellow labels, and the enhancing tumor core (ET) class is shown in yellow.
  • ADC Decreases in Solid Tumors Following Monotherapy With PEGylated Recombinant Human Hyaluronidase: Results From Early-Phase Clinical Trials
    Andres Mauricio Arias-Lorza1 and Natarajan Raghunand1
    1Moffitt Cancer Center, Tampa, FL, United States
    ADC decreases in solid tumors following monotherapy with PEGylated Recombinant Human Hyaluronidase PEGPH20.
    Figure 4. ADC changes in muscle tissue (top) and Bland-Altman plot of ADC repeats differences at muscle.
    Figure 3. Bland-Altman plot describing median ADC repeatability. Each marker/color represents a baseline repeat difference at each tumor. RC obtained per tumor is used to distinguish true changes in that tumor. In case the patient does not have more than one repeat at baseline, the full data RC given by the dashed lines is used instead.
  • Differential diagnosis of Endometrial carcinoma and polyps using Amide proton transfer-weighted imaging and permeability analysis
    Ye Li1, Xulun Lu1, Shifeng Tian1, Jiazheng Wang2, Zhiwei Shen2, Qingwei Song1, and Ailian Liu1
    1The First Affiliated Hospital of Dalian Medical University, Dalian, China, 2Philips Healthcare, Beijin, China
    APTw SI and permeability indictors based on DCE-MRI are potentially a promising and valuable non-invasive method in differentiation of endometrial carcinoma from endometrial polyps.
    table
    table
  • Preoperative discrimination between the low-grade glioma and high-grade glioma and early exploration of metastatic margin with APT imaging at 7T
    Yifan Yuan1, Qi Yue1, Xiang Zou1, Jiajun Cai1, Ying-Hua Chu2, Yi-Cheng Hsu2, Patrick Alexander Liebig3, Hui Zhang4, He Wang4, Liang Chen1, and Ying Mao1
    1Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China, 2MR Collaboration, Siemens Healthcare Ltd., Shanghai, China, 3Siemens Healthcare GmbH, Erlangen, Germany, 4Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
    We aim to establish a new grade-discrimination criterion via APT and explore the possible metastatic margin of diffuse glioma at 7T. With the early exploration of metastatic margin, the APT imaging at 7T may significantly facilitate the surgical strategy and surgical excision extension.
    Figure 1. T2-Flair, MP2RAGE, and ATP were acquired from Subject 9. The images in the top row is from the higher grade (WHO Grade IV) lesion. The results of the lower grade (WHO Grade II) lesion are shown in the bottom row. Larger APT values can be found in the high-grade region. Region of interest (ROI) was defined within the glioma lesion, and the statistical analysis was performed based on the ROIs.
    Figure 2. The group statistic using APT-CEST signal intensities obtained from all three grades of glioma were shown. (A) The APT-CEST signal intensity of high-grade (III and IV) glioma is significantly higher than low-grade (II) glioma (p-value <0.05). There was a significantly lower APT-CEST signal in WHO grade II compare to WHO grade IV (p-value <0.05). (B) The IDH mutation status in patient group was classified by WHO.
  • The Effect of Cramer-Rao Lower Bound Thresholds on Classification of IDH and TERTp Mutation Status in Gliomas using 1H-MRS
    Abdullah Bas1, Banu Sacli-Bilmez1, Ayca Ersen Danyeli2,3, Cengiz Yakicier3,4, M.Necmettin Pamir3,5, Koray Ozduman3,5, Alp Dincer3,6, and Esin Ozturk-Isik1,3
    1Institute of Biomedical Engineering, Bogazici University, İstanbul, Turkey, 2Department of Medical Pathology, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey, 3Center for Neuroradiological Applications and Reseach, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey, 4Department of Molecular Biology and Genetics, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey, 5Department of Neurosurgery, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey, 6Department of Radiology, Acıbadem Mehmet Ali Aydinlar University, Istanbul, Turkey
    CRLB values of GPC, PCh, 2HG, and Ins were different between the IDH and TERTp mutational subgroups of gliomas. Different CRLB thresholds followed by zero-imputing resulted in similar classification accuracies for IDH and TERTp mutations.
    Table 1: The p values obtained using a Mann-Whitney U test for assessing the CRLB differences between several mutational subgroups of gliomas. (*p<0.002, Bonferroni multiple comparison correction)
    Table 2: The classification results of the models for the detection of IDH and TERTp mutations based on different CRLB values. ‘No-FS’ means that all the metabolites were included in the classification.
  • Correlation between Amide Proton Transfer Imaging and Pathological Staging of Rectal Cancer
    Honglei Hu1, Xixi Zhao1, Qiming Wei2, Chuyao Chen1, Yuewei Huang1, Yingjie Mei3, and Yikai Xu1
    1Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou, China, 2Intervention, Traditional Chinese Medicine Hospital of Guangdong Province, Guangzhou, China, 3Philips Healthcare, Guangzhou, China
    The mean ATP SI(%) values at the tumor site of the largest cross-section of rectal cancer are closely related to its pathological stage, and they are statistically significant in pathological T stage and N stage(pT stage: p=0.008; pN Stage: p=0.000).
    Figure2. Magnetic resonance images showing a 57-year-old male patient with rectal cancer. The tumor is isointense on T1-weighted MR image, mildly hyperintense on T2-weighted MR image, hyperintense on DWI (b = 1000 s/mm2) , and isointense on ATP image (d).
    Table1. Correlations Between Mean ATP SI(%) with Clinicopathologic Characteristics.
  • Correlations of Single Voxel 1H-MRS Findings with Tumor Biology in Meningiomas.
    Banu Sacli-Bilmez1, Ayca Ersen Danyeli2,3, Murat Şakir Ekşi4, Kübra Tan5, Ozge Can6, Cengiz Yakicier7, M.Necmettin Pamir3,4, Alp Dincer3,8, Koray Özduman3,4, and Esin Ozturk-Isik1
    1Institute of Biomedical Engineering, Bogazici University, Istanbul, Turkey, 2Department of Medical Pathology, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey, 3Center for Neuroradiological Applications and Reseach, Acıbadem Mehmet Ali Aydinlar University, Istanbul, Turkey, 4Department of Neurosurgery, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey, 5Health Institutes of Turkey, Istanbul, Turkey, 6Department of Medical Engineering, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey, 7Department of Molecular Biology and Genetics, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey, 8Department of Radiology, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
    Higher Glyc and Ins+Glyc levels in aggressive meningiomas, and higher Glu, GPC, and Glx levels in hyperostotic tumors as determined by noninvasive, clinical, single-voxel 1H-MRS can provide clinically useful clues on tumor biology in meningiomas.
    Figure 1. MR spectroscopic data with some of the LCModel results of grade I (a) and grade II (b) meningioma.
    Table 1. The metabolite peak intensity differences between different grades of meningiomas and the p-values of a Mann-Whitney U test.
  • Early detection of radiation-induced injury and prediction of cognitive deficit by MRS metabolites in radiotherapy of low grade glioma
    Zahra Alirezaei1,2, Mohammadreza Nazemzadeh3, Masoud Hasanpour3, Alireza Amouheidari4, and Sajad Iraji3
    1Isfahan University of Medical Sciences, Isfahan, Iran (Islamic Republic of), 2Bushehr University of Medical Sciences, Bushehr, Iran (Islamic Republic of), 3Tehran University of Medical Sciences, Tehran, Iran (Islamic Republic of), 4Milad Hospital, Isfahan, Iran (Islamic Republic of)
    NAA/Cr and Cho/Cr declined significantly at the 4th week of RT up to 6-month post-RT. The variation in NAA/Cr and Cho/Cr between the 4th week of RT and 1-month post-RT had significant correlation with the alteration in the score of MoCA-visuospatial and ACE-memory, between 3 and 6-month post-RT. 
    Figure 1. a: Isodose curves of a treatment plan of a LGG patient with a Left Frontal tumor on Prowess Panther 5.5; b: An example of MRS metabolite peaks of a voxel on the CC, fused with T2W MRI on a 1.5 Tesla Siemens Magnetom Aera scanner.
    Figure 2. Plots of the mean percantage difference in the mean value of NAA/Cr and Cho/Cralong with the ACE and MoCA Scores at the 4th week of RT, 1-month, 3-month and 6-month post-RT, compared to the previous timepoints and baseline pre-RT values. The star markers show significant differences to the base line, while the plus ones present the significant differences between any parameters to its previous time point.
  • 31P spectral profiles in brain tissues of volunteers and glioma patients at 7T
    Andreas Korzowski1, Nina Weckesser2, Vanessa L Franke1, Heinz-Peter Schlemmer2, Mark E Ladd1, Peter Bachert1, and Daniel Paech2
    1Medical Physics in Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany, 2Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
    The presented high-quality 31P brain spectra from individual tissue types obtained at B0 = 7T illustrate clear differences not only between healthy and tumor tissues, but also between different compartments within diseased tissue.
    Subject-averaged 31P spectra (black lines) of individual brain tissue types, with corresponding standard deviation across subjects (gray shading). For white matter (A), 15 ROIs were averaged (6 from volunteers, 9 from patients), and 3 ROIs for gray matter (B). For edema (C), 9 ROIs were averaged (from all patients), and 7 ROIs for Gd-contrast enhancement (D, from all high-grade glioma patients).
    Example data processing for a GDCE ROI in a high-grade glioma patient. The high-resolution ROI (green) drawn on 1H images is mapped onto the interpolated 31P MRSI grid (matrix = 80×96×64, 31P intensity map is shown). Within the resulting low-resolution ROI (blue), phase-/frequency-aligned spectra are summed up to yield the ROI-averaged spectrum. PCr, phosphocreatine; ATP, adenosine-5’-triphosphate; NAD(H), nicotinamide dinucleotide, UDPG, uridine disphosphoglucose; (G)PE, (glycero)phosphoethanolamine; (G)PC, (glycero)phosphocholine; Pi, inorganic phosphate.
  • 1D-CNN for the Detection of IDH and TERTp Mutations in Diffuse Gliomas using Proton Magnetic Resonance Spectroscopy
    Abdullah BAS1, Banu Sacli-Bilmez1, Ayca Ersen Danyeli2,3, Cengiz Yakicier3,4, M.Necmettin Pamir3,5, Koray Ozduman3,5, Alp Dincer3,6, and Esin Ozturk-Isik1,3
    1Institute of Biomedical Engineering, Bogazici University, Istanbul, Turkey, 2Department of Medical Pathology, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey, 3Center for Neuroradiological Applications and Reseach, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey, 4Department of Molecular Biology and Genetics, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey, 5Department of Neurosurgery, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey, 6Department of Radiology, Acıbadem Mehmet Ali Aydinlar University, Istanbul, Turkey
    This study indicated that 1D-CNN models could identify IDH-mut, TERTp-mut and TERTp-only gliomas using 1H-MRS with 94.11%, 76.92%, and 82.05% accuracies, respectively. Deep-learning techniques might be promising for the mutational classification of 1H-MRS data of gliomas.
    Table 2. The performance metrics of the 1D-CNN models on test and validation set.
    Figure 2. Example short TE PRESS 1H-MRS data for (a) IDH-mut&TERTp-mut, (b) IDH-mut&TERTp-wt, (c) TERTp-only, and (d) IDH-wt&TERTp-wt gliomas.
  • Metabolic profiles of glioma grade and IDH mutation status using high resolution 7T 3D-FID-MRSI: Preliminary results
    Cornelius Cadrien1,2, Sukrit Sharma1, Philipp Lazen1, Julia Furtner3, Alexandra Lipka1,4, Eva Hečková1, Lukas Hingerl1, Stanislav Motyka1, Stephan Gruber1, Bernhard Strasser1, Barbara Kiesel2, Mario Mischkulnig2, Matthias Preusser5, Thomas Roetzer6, Adelheid Wöhrer6, Michael Weber7, Christian Dorfer2, Karl Rössler2, Siegfried Trattnig1,4, Wolfgang Bogner1, Georg Widhalm2, and Gilbert Hangel1,2
    1High-field MR Center, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria, 2Department of Neurosurgery, Medical University of Vienna, Vienna, Austria, 3Division of Neuroradiology and Musculoskeletal Radiology, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria, 4Christian Doppler Laboratory for Clinical Molecular MR Imaging, Vienna, Austria, 5Division of Oncology, Department of Inner Medicine I, Medical University of Vienna, Vienna, Austria, 6Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria, 7Division of Medical Imaging and Nuclear Medicine, Medical University of Vienna, Vienna, Austria
    We investigated 7T MRSI- based metabolite ratio maps of 36 WHO grade 2-4 glioma patients in defined ROIs. We found significant differences between tumor and NAWM ROIs as well as significantly less mIns/tCr in high- versus low-grades.

    Fig 3 - Direct comparison figure - LGG vs HGG

    We can see ratio maps of different metabolites to Creatine ratios and the 3T contrast-enhancing clinical images, as well as the 7T T1 images. The LGG patient on top shows a different metabolite pattern in the region of the contrast-enhancing region, compared to the HGG patient below.

    Fig 2 - Boxplots tumor ROI vs WM ROI

    Most of the metabolites show significant differences in median voxel values of the tumor hotspot region vs. NAWM (see. A). On the B side, we can see a sample hotspot map of tCho with applied cutoff of 0.36 and the 7T T1 magnitude.