1
|
Moloney B, Li X, Hirano M, Saad Eddin A, Lim JY, Biswas D, Kazerouni AS, Tudorica A, Li I, Bryant ML, Wille C, Pyle C, Rahbar H, Hsieh SK, Rice-Stitt TL, Dintzis SM, Bashir A, Hobbs E, Zimmer A, Specht JM, Phadke S, Fleege N, Holmes JH, Partridge SC, Huang W. Initial experience in implementing quantitative DCE-MRI to predict breast cancer therapy response in a multi-center and multi-vendor platform setting. Front Oncol 2024; 14:1395502. [PMID: 39678499 PMCID: PMC11638047 DOI: 10.3389/fonc.2024.1395502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 10/28/2024] [Indexed: 12/17/2024] Open
Abstract
Quantitative dynamic contrast-enhanced (DCE) MRI as a promising method for the prediction of breast cancer response to neoadjuvant chemotherapy (NAC) has been demonstrated mostly in single-center and single-vendor platform studies. This preliminary study reports the initial experience in implementing quantitative breast DCE-MRI in multi-center (MC) and multi-vendor platform (MP) settings to predict NAC response. MRI data, including B1 mapping, variable flip angle (VFA) measurements of native tissue R1 (R1,0), and DCE-MRI, were acquired during NAC at three sites using 3T systems with Siemens, Philips, and GE platforms, respectively. High spatiotemporal resolution DCE-MRI was performed using similar vendor product sequences with k-space undersampling during acquisition and view sharing during reconstruction. A breast phantom was used for quality assurance/quality control (QA/QC) across sites. The Tofts model (TM) and shutter-speed model (SSM) were used for pharmacokinetic (PK) analysis of the DCE data. Additionally, tumor region of interest (ROI)- vs. voxel-based analyses in combination with the use of VFA-measured R1,0 vs. fixed, literature-reported R1,0 were investigated to determine the optimal analysis approach. Results from 15 patients who completed the study are reported. Voxel-based PK analysis using fixed R1,0 was deemed the optimal approach, which allowed the inclusion of data from one vendor platform where VFA measurements produced ≥100% overestimation of R1,0. The semi-quantitative signal enhancement ratio (SER) and quantitative PK parameters outperformed the tumor longest diameter (LD) in the prediction of pathologic complete response (pCR) vs. non-pCR after the first NAC cycle, whereas Ktrans consistently provided more accurate predictions than both SER and LD after the first NAC cycle and at the NAC midpoint. Both TM and SSM Ktrans and kep were excellent predictors of response at the NAC midpoint with ROC AUC >0.90, while the SSM parameters (AUC ≥0.80) performed better than their TM counterparts (AUC <0.80) after the first NAC cycle. The initial experience of this ongoing study indicates the importance of QA/QC using a phantom and suggests that deploying voxel-based PK analysis using a fixed R1,0 may mitigate random errors from R1,0 measurements across platforms and potentially eliminate the need for B1 and VFA acquisitions in MC and MP trials.
Collapse
Affiliation(s)
- Brendan Moloney
- Advanced Imaging Research Center, Oregon Health and Science University, Portland, OR, United States
| | - Xin Li
- Advanced Imaging Research Center, Oregon Health and Science University, Portland, OR, United States
| | - Michael Hirano
- Department of Radiology, University of Washington, Seattle, WA, United States
| | - Assim Saad Eddin
- Department of Radiology, University of Iowa, Iowa City, IA, United States
| | - Jeong Youn Lim
- Biostatistics Shared Resource, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, United States
| | - Debosmita Biswas
- Department of Radiology, University of Washington, Seattle, WA, United States
| | - Anum S. Kazerouni
- Department of Radiology, University of Washington, Seattle, WA, United States
| | - Alina Tudorica
- Department of Diagnostic Radiology, Oregon Health and Science University, Portland, OR, United States
| | - Isabella Li
- Department of Radiology, University of Washington, Seattle, WA, United States
| | - Mary Lynn Bryant
- Department of Radiology, University of Washington, Seattle, WA, United States
| | - Courtney Wille
- Institute for Clinical and Translational Science, University of Iowa, Iowa City, IA, United States
| | - Chelsea Pyle
- Department of Diagnostic Radiology, Oregon Health and Science University, Portland, OR, United States
| | - Habib Rahbar
- Department of Radiology, University of Washington, Seattle, WA, United States
- Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Su Kim Hsieh
- Department of Radiology, University of Iowa, Iowa City, IA, United States
| | - Travis L. Rice-Stitt
- Department of Pathology, Oregon Health and Science University, Portland, OR, United States
| | - Suzanne M. Dintzis
- Fred Hutchinson Cancer Center, Seattle, WA, United States
- Department of Pathology, University of Washington, Seattle, WA, United States
| | - Amani Bashir
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, United States
- Department of Pathology, University of Iowa Hospitals and Clinics, Iowa City, IA, United States
| | - Evthokia Hobbs
- Hematology and Medical Oncology Division, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, United States
| | - Alexandra Zimmer
- Hematology and Medical Oncology Division, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, United States
| | - Jennifer M. Specht
- Fred Hutchinson Cancer Center, Seattle, WA, United States
- Division of Hematology and Oncology, University of Washington, Seattle, WA, United States
| | - Sneha Phadke
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, United States
- Department of Internal Medicine, University of Iowa Hospitals and Clinics, Iowa City, IA, United States
| | - Nicole Fleege
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, United States
- Department of Internal Medicine, University of Iowa Hospitals and Clinics, Iowa City, IA, United States
| | - James H. Holmes
- Department of Radiology, University of Iowa, Iowa City, IA, United States
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, United States
| | - Savannah C. Partridge
- Department of Radiology, University of Washington, Seattle, WA, United States
- Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Wei Huang
- Advanced Imaging Research Center, Oregon Health and Science University, Portland, OR, United States
| |
Collapse
|
2
|
Arledge CA, Zhao AH, Topaloglu U, Zhao D. Dynamic Contrast Enhanced MRI Mapping of Vascular Permeability for Evaluation of Breast Cancer Neoadjuvant Chemotherapy Response Using Image-to-Image Conditional Generative Adversarial Networks. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.04.24313070. [PMID: 39281733 PMCID: PMC11398591 DOI: 10.1101/2024.09.04.24313070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/18/2024]
Abstract
Dynamic contrast enhanced (DCE) MRI is a non-invasive imaging technique that has become a quantitative standard for assessing tumor microvascular permeability. Through the application of a pharmacokinetic (PK) model to a series of T1-weighed MR images acquired after an injection of a contrast agent, several vascular permeability parameters can be quantitatively estimated. These parameters, including Ktrans, a measure of capillary permeability, have been widely implemented for assessing tumor vascular function as well as tumor therapeutic response. However, conventional PK modeling for translation of DCE MRI to PK vascular permeability parameter maps is complex and time-consuming for dynamic scans with thousands of pixels per image. In recent years, image-to-image conditional generative adversarial network (cGAN) is emerging as a robust approach in computer vision for complex cross-domain translation tasks. Through a sophisticated adversarial training process between two neural networks, image-to-image cGANs learn to effectively translate images from one domain to another, producing images that are indistinguishable from those in the target domain. In the present study, we have developed a novel image-to-image cGAN approach for mapping DCE MRI data to PK vascular permeability parameter maps. The DCE-to-PK cGAN not only generates high-quality parameter maps that closely resemble the ground truth, but also significantly reduces computation time over 1000-fold. The utility of the cGAN approach to map vascular permeability is validated using open-source breast cancer patient DCE MRI data provided by The Cancer Imaging Archive (TCIA). This data collection includes images and pathological analyses of breast cancer patients acquired before and after the first cycle of neoadjuvant chemotherapy (NACT). Importantly, in good agreement with previous studies leveraging this dataset, the percentage change of vascular permeability Ktrans derived from the DCE-to-PK cGAN enables early prediction of responders to NACT.
Collapse
Affiliation(s)
- Chad A. Arledge
- Department of Biomedical Engineering, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Alan H. Zhao
- University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Umit Topaloglu
- Clinical and Translational Research Informatics Branch, National Cancer Institute, Rockville, MD 20850, USA
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Dawen Zhao
- Department of Biomedical Engineering, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| |
Collapse
|
3
|
Rastogi A, Yalavarthy PK. Greybox: A hybrid algorithm for direct estimation of tracer kinetic parameters from undersampled DCE-MRI data. Med Phys 2024; 51:4838-4858. [PMID: 38214325 DOI: 10.1002/mp.16935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 11/28/2023] [Accepted: 12/22/2023] [Indexed: 01/13/2024] Open
Abstract
BACKGROUND A variety of deep learning-based and iterative approaches are available to predict Tracer Kinetic (TK) parameters from fully sampled or undersampled dynamic contrast-enhanced (DCE) MRI data. However, both the methods offer distinct benefits and drawbacks. PURPOSE To propose a hybrid algorithm (named as 'Greybox'), using both model- as well as DL-based, for solving a multi-parametric non-linear inverse problem of directly estimating TK parameters from undersampled DCE MRI data, which is invariant to undersampling rate. METHODS The proposed algorithm was inspired by plug-and-play algorithms used for solving linear inverse imaging problems. This technique was tested for its effectiveness in solving the nonlinear ill-posed inverse problem of generating 3D TK parameter maps from four-dimensional (4D; Spatial + Temporal) retrospectively undersampled k-space data. The algorithm learns a deep learning-based prior using UNET to estimate theK trans $\mathbf {K_{trans}}$ andV p $\mathbf {V_{p}}$ parameters based on the Patlak pharmacokinetic model, and this trained prior was utilized to estimate the TK parameter maps using an iterative gradient-based optimization scheme. Unlike the existing DL models, this network is invariant to the undersampling rate of the input data. The proposed method was compared with the total variation-based direct reconstruction technique on brain, breast, and prostate DCE-MRI datasets for various undersampling rates using the Radial Golden Angle (RGA) scheme. For the breast dataset, an indirect estimation using the Fast Composite Splitting algorithm was utilized for comparison. Undersampling rates of 8 × $\times$ , 12 × $\times$ and 20 × $\times$ were used for the experiments, and the results were compared using the PSNR and SSIM as metrics. For the breast dataset of 10 patients, data from four patients were utilized for training (1032 samples), two for validation (752 samples), and the entire volume of four patients for testing. Similarly, for the prostate dataset of 18 patients, 10 patients were utilized for training (720 samples), five for validation (216 samples), and the whole volume of three patients for testing. For the brain dataset of nineteen patients, ten patients were used for training (3152 samples), five for validation (1168 samples), and the whole volume of four patients for testing. Statistical tests were also conducted to assess the significance of the improvement in performance. RESULTS The experiments showed that the proposed Greybox performs significantly better than other direct reconstruction methods. The proposed algorithm improved the estimatedK trans $\mathbf {K_{trans}}$ andV p $\mathbf {V_{p}}$ in terms of the peak signal-to-noise ratio by up to 3 dB compared to other standard reconstruction methods. CONCLUSION The proposed hybrid reconstruction algorithm, Greybox, can provide state-of-the-art performance in solving the nonlinear inverse problem of DCE-MRI. This is also the first of its kind to utilize convolutional neural network-based encodings as part of the plug-and-play priors to improve the performance of the reconstruction algorithm.
Collapse
Affiliation(s)
- Aditya Rastogi
- Department of Computational and Data Sciences, Indian Institute of Science, Bangalore, India
- University Hospital Heidelberg, Heidelberg, Germany
| | | |
Collapse
|
4
|
Dickie BR, Ahmed Z, Arvidsson J, Bell LC, Buckley DL, Debus C, Fedorov A, Floca R, Gutmann I, van der Heijden RA, van Houdt PJ, Sourbron S, Thrippleton MJ, Quarles C, Kompan IN. A community-endorsed open-source lexicon for contrast agent-based perfusion MRI: A consensus guidelines report from the ISMRM Open Science Initiative for Perfusion Imaging (OSIPI). Magn Reson Med 2024; 91:1761-1773. [PMID: 37831600 PMCID: PMC11337559 DOI: 10.1002/mrm.29840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 07/25/2023] [Accepted: 08/04/2023] [Indexed: 10/15/2023]
Abstract
This manuscript describes the ISMRM OSIPI (Open Science Initiative for Perfusion Imaging) lexicon for dynamic contrast-enhanced and dynamic susceptibility-contrast MRI. The lexicon was developed by Taskforce 4.2 of OSIPI to provide standardized definitions of commonly used quantities, models, and analysis processes with the aim of reducing reporting variability. The taskforce was established in February 2020 and consists of medical physicists, engineers, clinicians, data and computer scientists, and DICOM (Digital Imaging and Communications in Medicine) standard experts. Members of the taskforce collaborated via a slack channel and quarterly virtual meetings. Members participated by defining lexicon items and reporting formats that were reviewed by at least two other members of the taskforce. Version 1.0.0 of the lexicon was subject to open review from the wider perfusion imaging community between January and March 2022, and endorsed by the Perfusion Study Group of the ISMRM in the summer of 2022. The initial scope of the lexicon was set by the taskforce and defined such that it contained a basic set of quantities, processes, and models to enable users to report an end-to-end analysis pipeline including kinetic model fitting. We also provide guidance on how to easily incorporate lexicon items and definitions into free-text descriptions (e.g., in manuscripts and other documentation) and introduce an XML-based pipeline encoding format to encode analyses using lexicon definitions in standardized and extensible machine-readable code. The lexicon is designed to be open-source and extendable, enabling ongoing expansion of its content. We hope that widespread adoption of lexicon terminology and reporting formats described herein will increase reproducibility within the field.
Collapse
Affiliation(s)
- Ben R. Dickie
- Division of Informatics, Imaging, and Data Sciences, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- Geoffrey Jefferson Brain Research Center, Manchester Academic Health Science Center, The University of Manchester, Manchester, UK
| | - Zaki Ahmed
- Corewell Health William Beaumont University Hospital, Royal Oak, Michigan, USA
| | - Jonathan Arvidsson
- Department of Medical Radiation Sciences, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Medical Physics and Biomedical Engineering, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Laura C. Bell
- Clinical Imaging Group, Genentech, Inc., South San Francisco, California, USA
| | | | | | - Andrey Fedorov
- Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ralf Floca
- National Center for Radiation Research in Oncology, Heidelberg Institute for Radiation Oncology, Heidelberg, Germany
| | - Ingomar Gutmann
- Faculty of Physics, Physics of Functional Materials, University of Vienna, Vienna, Austria
| | - Rianne A. van der Heijden
- Department of Radiology & Nuclear Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Department of Radiology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Petra J. van Houdt
- Department of Radiation Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Steven Sourbron
- Department of Infection, Immunity, and Cardiovascular Diseases, University of Sheffield, Sheffield, UK
| | - Michael J. Thrippleton
- Edinburgh Imaging and Center for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Chad Quarles
- Department of Cancer Systems Imaging, UT MD Anderson Cancer Center, Houston, Texas, USA
| | - Ina N. Kompan
- Division of Medical Image Computing, German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
5
|
van Houdt PJ, Ragunathan S, Berks M, Ahmed Z, Kershaw LE, Gurney-Champion OJ, Tadimalla S, Arvidsson J, Sun Y, Kallehauge J, Dickie B, Lévy S, Bell L, Sourbron S, Thrippleton MJ. Contrast-agent-based perfusion MRI code repository and testing framework: ISMRM Open Science Initiative for Perfusion Imaging (OSIPI). Magn Reson Med 2024; 91:1774-1786. [PMID: 37667526 DOI: 10.1002/mrm.29826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 06/30/2023] [Accepted: 07/25/2023] [Indexed: 09/06/2023]
Abstract
PURPOSE Software has a substantial impact on quantitative perfusion MRI values. The lack of generally accepted implementations, code sharing and transparent testing reduces reproducibility, hindering the use of perfusion MRI in clinical trials. To address these issues, the ISMRM Open Science Initiative for Perfusion Imaging (OSIPI) aimed to establish a community-led, centralized repository for sharing open-source code for processing contrast-based perfusion imaging, incorporating an open-source testing framework. METHODS A repository was established on the OSIPI GitHub website. Python was chosen as the target software language. Calls for code contributions were made to OSIPI members, the ISMRM Perfusion Study Group, and publicly via OSIPI websites. An automated unit-testing framework was implemented to evaluate the output of code contributions, including visual representation of the results. RESULTS The repository hosts 86 implementations of perfusion processing steps contributed by 12 individuals or teams. These cover all core aspects of DCE- and DSC-MRI processing, including multiple implementations of the same functionality. Tests were developed for 52 implementations, covering five analysis steps. For T1 mapping, signal-to-concentration conversion and population AIF functions, different implementations resulted in near-identical output values. For the five pharmacokinetic models tested (Tofts, extended Tofts-Kety, Patlak, two-compartment exchange, and two-compartment uptake), differences in output parameters were observed between contributions. CONCLUSIONS The OSIPI DCE-DSC code repository represents a novel community-led model for code sharing and testing. The repository facilitates the re-use of existing code and the benchmarking of new code, promoting enhanced reproducibility in quantitative perfusion imaging.
Collapse
Affiliation(s)
- Petra J van Houdt
- Department of Radiation Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - Michael Berks
- Quantitative Biomedical Imaging Laboratory, Division of Cancer Sciences, The University of Manchester, Manchester, UK
| | - Zaki Ahmed
- Corewell Health William Beaumont University Hospital, Diagnostic Radiology, Royal Oak, USA
| | - Lucy E Kershaw
- Edinburgh Imaging and Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Oliver J Gurney-Champion
- Department of Radiology and Nuclear Medicine, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Sirisha Tadimalla
- Institute of Medical Physics, The University of Sydney, Sydney, Australia
| | - Jonathan Arvidsson
- Department of Medical Radiation Sciences, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Medical Physics and Biomedical Engineering, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Yu Sun
- Institute of Medical Physics, The University of Sydney, Sydney, Australia
| | - Jesper Kallehauge
- Aarhus University Hospital, Danish Centre for Particle Therapy, Aarhus, Denmark
- Aarhus University, Department of Clinical Medicine, Aarhus, Denmark
| | - Ben Dickie
- Division of Informatics, Imaging, and Data Science, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Group, The University of Manchester, Manchester, UK
| | - Simon Lévy
- MR Research Collaborations, Siemens Healthcare Pty Ltd, Melbourne, Australia
| | - Laura Bell
- Genentech, Inc, Clinical Imaging Group, South San Francisco, USA
| | - Steven Sourbron
- University of Sheffield, Department of Infection, Immunity and Cardiovascular Disease, Sheffield, UK
| | - Michael J Thrippleton
- University of Edinburgh, Edinburgh Imaging and Centre for Clinical Brain Sciences, Edinburgh, UK
| |
Collapse
|
6
|
Abstract
The non-invasive dynamic contrast-enhanced MRI (DCE-MRI) method provides valuable insights into tissue perfusion and vascularity. Primarily used in oncology, DCE-MRI is typically utilized to assess morphology and contrast agent (CA) kinetics in the tissue of interest. Interpretation of the temporal signatures of DCE-MRI data includes qualitative, semi-quantitative, and quantitative approaches. Recent advances in MRI technology allow simultaneous high spatial and temporal resolutions in DCE-MRI data acquisition on most vendor platforms, enabling the more desirable approach of quantitative data analysis using pharmacokinetic (PK) modeling. Many technical factors, including signal-to-noise ratio, temporal resolution, quantifications of arterial input function and native tissue T1, and PK model selection, need to be carefully considered when performing quantitative DCE-MRI. Standardization in data acquisition and analysis is especially important in multi-center studies.
Collapse
Affiliation(s)
- Xin Li
- Advanced Imaging Research Center, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Wei Huang
- Advanced Imaging Research Center, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - James H Holmes
- Radiology, Biomedical Engineering, and Holden Cancer Center, University of Iowa, 169 Newton Road, Iowa City, IA 52242, USA.
| |
Collapse
|
7
|
Zhao Z, Du S, Xu Z, Yin Z, Huang X, Huang X, Wong C, Liang Y, Shen J, Wu J, Qu J, Zhang L, Cui Y, Wang Y, Wee L, Dekker A, Han C, Liu Z, Shi Z, Liang C. SwinHR: Hemodynamic-powered hierarchical vision transformer for breast tumor segmentation. Comput Biol Med 2024; 169:107939. [PMID: 38194781 DOI: 10.1016/j.compbiomed.2024.107939] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 12/12/2023] [Accepted: 01/01/2024] [Indexed: 01/11/2024]
Abstract
Accurate and automated segmentation of breast tumors in dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) plays a critical role in computer-aided diagnosis and treatment of breast cancer. However, this task is challenging, due to random variation in tumor sizes, shapes, appearances, and blurred boundaries of tumors caused by inherent heterogeneity of breast cancer. Moreover, the presence of ill-posed artifacts in DCE-MRI further complicate the process of tumor region annotation. To address the challenges above, we propose a scheme (named SwinHR) integrating prior DCE-MRI knowledge and temporal-spatial information of breast tumors. The prior DCE-MRI knowledge refers to hemodynamic information extracted from multiple DCE-MRI phases, which can provide pharmacokinetics information to describe metabolic changes of the tumor cells over the scanning time. The Swin Transformer with hierarchical re-parameterization large kernel architecture (H-RLK) can capture long-range dependencies within DCE-MRI while maintaining computational efficiency by a shifted window-based self-attention mechanism. The use of H-RLK can extract high-level features with a wider receptive field, which can make the model capture contextual information at different levels of abstraction. Extensive experiments are conducted in large-scale datasets to validate the effectiveness of our proposed SwinHR scheme, demonstrating its superiority over recent state-of-the-art segmentation methods. Also, a subgroup analysis split by MRI scanners, field strength, and tumor size is conducted to verify its generalization. The source code is released on (https://github.com/GDPHMediaLab/SwinHR).
Collapse
Affiliation(s)
- Zhihe Zhao
- School of Medicine, South China University of Technology, Guangzhou, 510006, China; Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China; Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, 510080, China
| | - Siyao Du
- Department of Radiology, The First Hospital of China Medical University, Shenyang, Liaoning Province, 110001, China
| | - Zeyan Xu
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, 650118, China
| | - Zhi Yin
- Department of Radiology, Shanxi Province Cancer Hospital/ Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030013, China
| | - Xiaomei Huang
- Department of Medical Imaging, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xin Huang
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China; Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, 510080, China; Shantou University Medical College, Shantou, 515041, China
| | - Chinting Wong
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China; Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, 510080, China
| | - Yanting Liang
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China; Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, 510080, China
| | - Jing Shen
- Department of Radiology, Affiliated Zhongshan Hospital of Dalian University, Dalian, 116001, China
| | - Jianlin Wu
- Department of Radiology, Affiliated Zhongshan Hospital of Dalian University, Dalian, 116001, China
| | - Jinrong Qu
- Department of Radiology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Lina Zhang
- Department of Radiology, The First Hospital of China Medical University, Shenyang, Liaoning Province, 110001, China
| | - Yanfen Cui
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, 510080, China; Department of Radiology, Shanxi Province Cancer Hospital/ Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030013, China
| | - Ying Wang
- Department of Medical Ultrasonics, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Leonard Wee
- Clinical Data Science, Faculty of Health Medicine Life Sciences, Maastricht University, Maastricht, 6229 ET, The Netherlands; Department of Radiation Oncology (Maastro), GROW School of Oncology and Reproduction, Maastricht University Medical Centre+, Maastricht, 6229 ET, The Netherlands
| | - Andre Dekker
- Department of Radiation Oncology (Maastro), GROW School of Oncology and Reproduction, Maastricht University Medical Centre+, Maastricht, 6229 ET, The Netherlands
| | - Chu Han
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China; Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, 510080, China; Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
| | - Zaiyi Liu
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China; Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, 510080, China.
| | - Zhenwei Shi
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China; Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, 510080, China; Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
| | - Changhong Liang
- School of Medicine, South China University of Technology, Guangzhou, 510006, China; Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China; Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, 510080, China.
| |
Collapse
|
8
|
Stoyanova R, Zavala-Romero O, Kwon D, Breto AL, Xu IR, Algohary A, Alhusseini M, Gaston SM, Castillo P, Kryvenko ON, Davicioni E, Nahar B, Spieler B, Abramowitz MC, Dal Pra A, Parekh DJ, Punnen S, Pollack A. Clinical-Genomic Risk Group Classification of Suspicious Lesions on Prostate Multiparametric-MRI. Cancers (Basel) 2023; 15:5240. [PMID: 37958414 PMCID: PMC10647832 DOI: 10.3390/cancers15215240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/12/2023] [Accepted: 10/24/2023] [Indexed: 11/15/2023] Open
Abstract
The utilization of multi-parametric MRI (mpMRI) in clinical decisions regarding prostate cancer patients' management has recently increased. After biopsy, clinicians can assess risk using National Comprehensive Cancer Network (NCCN) risk stratification schema and commercially available genomic classifiers, such as Decipher. We built radiomics-based models to predict lesions/patients at low risk prior to biopsy based on an established three-tier clinical-genomic classification system. Radiomic features were extracted from regions of positive biopsies and Normally Appearing Tissues (NAT) on T2-weighted and Diffusion-weighted Imaging. Using only clinical information available prior to biopsy, five models for predicting low-risk lesions/patients were evaluated, based on: 1: Clinical variables; 2: Lesion-based radiomic features; 3: Lesion and NAT radiomics; 4: Clinical and lesion-based radiomics; and 5: Clinical, lesion and NAT radiomic features. Eighty-three mpMRI exams from 78 men were analyzed. Models 1 and 2 performed similarly (Area under the receiver operating characteristic curve were 0.835 and 0.838, respectively), but radiomics significantly improved the lesion-based performance of the model in a subset analysis of patients with a negative Digital Rectal Exam (DRE). Adding normal tissue radiomics significantly improved the performance in all cases. Similar patterns were observed on patient-level models. To the best of our knowledge, this is the first study to demonstrate that machine learning radiomics-based models can predict patients' risk using combined clinical-genomic classification.
Collapse
Affiliation(s)
- Radka Stoyanova
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA
| | - Olmo Zavala-Romero
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Deukwoo Kwon
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Adrian L. Breto
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Isaac R. Xu
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Ahmad Algohary
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Mohammad Alhusseini
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Sandra M. Gaston
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA
| | - Patricia Castillo
- Department of Radiology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Oleksandr N. Kryvenko
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Desai Sethi Urology Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Elai Davicioni
- Research and Development, Veracyte Inc., San Francisco, CA 94080, USA
| | - Bruno Nahar
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA
- Desai Sethi Urology Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Benjamin Spieler
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA
| | - Matthew C. Abramowitz
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA
| | - Alan Dal Pra
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA
| | - Dipen J. Parekh
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA
- Desai Sethi Urology Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Sanoj Punnen
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA
- Desai Sethi Urology Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Alan Pollack
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA
| |
Collapse
|
9
|
Gill AB, Gallagher FA, Graves MJ. Open source code for the generation of digital reference objects for dynamic contrast-enhanced MRI analysis software validation. Br J Radiol 2023; 96:20220976. [PMID: 37191274 PMCID: PMC10321261 DOI: 10.1259/bjr.20220976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 03/01/2023] [Accepted: 03/29/2023] [Indexed: 05/17/2023] Open
Abstract
OBJECTIVES Dynamic contrast-enhanced MR images can be analyzed through the application of a wide range of kinetic models. This process is prone to variability and a lack of standardization that can affect the measured metrics. There is a need for customized digital reference objects (DROs) for the validation of DCE-MRI software packages that undertake kinetic model analysis. DROs are currently available only for a small subset of the kinetic models commonly applied to DCE-MRI data. This work aimed to address this gap. METHODS Code was written in the MATLAB programming environment to generate customizable DROs. This modular code allows the insertion of a plug-in to describe the kinetic model to be tested. We input our generated DROs into three commercial and open-source analysis packages and assessed the agreement of kinetic model parameter values output with the 'ground-truth' values used in the DRO generation. RESULTS For the five kinetic models tested, the concordance correlation coefficient values were >98%, indicating excellent agreement of the results with 'ground-truth'. CONCLUSIONS Testing our DROs on three independent software packages produced concordant results, strongly suggesting our DRO generation code is correct. This implies that our DROs can be used to validate other third party software for the kinetic model analysis of DCE-MRI data. ADVANCES IN KNOWLEDGE This work extends published work of others to allow customized generation of test objects for any applied kinetic model and allows the incorporation of B1 mapping into the DRO for application at higher field strengths.
Collapse
Affiliation(s)
- Andrew B. Gill
- Department of Radiology, University of Cambridge, Cambridge, UK
| | | | | |
Collapse
|
10
|
Ru J, Lu B, Chen B, Shi J, Chen G, Wang M, Pan Z, Lin Y, Gao Z, Zhou J, Liu X, Zhang C. Attention guided neural ODE network for breast tumor segmentation in medical images. Comput Biol Med 2023; 159:106884. [PMID: 37071938 DOI: 10.1016/j.compbiomed.2023.106884] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 01/25/2023] [Accepted: 03/30/2023] [Indexed: 04/05/2023]
Abstract
Breast cancer is the most common cancer in women. Ultrasound is a widely used screening tool for its portability and easy operation, and DCE-MRI can highlight the lesions more clearly and reveal the characteristics of tumors. They are both noninvasive and nonradiative for assessment of breast cancer. Doctors make diagnoses and further instructions through the sizes, shapes and textures of the breast masses showed on medical images, so automatic tumor segmentation via deep neural networks can to some extent assist doctors. Compared to some challenges which the popular deep neural networks have faced, such as large amounts of parameters, lack of interpretability, overfitting problem, etc., we propose a segmentation network named Att-U-Node which uses attention modules to guide a neural ODE-based framework, trying to alleviate the problems mentioned above. Specifically, the network uses ODE blocks to make up an encoder-decoder structure, feature modeling by neural ODE is completed at each level. Besides, we propose to use an attention module to calculate the coefficient and generate a much refined attention feature for skip connection. Three public available breast ultrasound image datasets (i.e. BUSI, BUS and OASBUD) and a private breast DCE-MRI dataset are used to assess the efficiency of the proposed model, besides, we upgrade the model to 3D for tumor segmentation with the data selected from Public QIN Breast DCE-MRI. The experiments show that the proposed model achieves competitive results compared with the related methods while mitigates the common problems of deep neural networks.
Collapse
Affiliation(s)
- Jintao Ru
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People's Republic of China
| | - Beichen Lu
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People's Republic of China
| | - Buran Chen
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People's Republic of China
| | - Jialin Shi
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People's Republic of China
| | - Gaoxiang Chen
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People's Republic of China
| | - Meihao Wang
- Department of Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People's Republic of China; Key Laboratory of Intelligent Medical Imaging of Wenzhou, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People's Republic of China.
| | - Zhifang Pan
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People's Republic of China; Zhejiang Engineering Research Center of Intelligent Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People's Republic of China.
| | - Yezhi Lin
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People's Republic of China; Key Laboratory of Intelligent Treatment and Life Support for Critical Diseases of Zhejiang Province, Wenzhou, 325000, People's Republic of China.
| | - Zhihong Gao
- Zhejiang Engineering Research Center of Intelligent Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People's Republic of China
| | - Jiejie Zhou
- Department of Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People's Republic of China
| | - Xiaoming Liu
- School of Computer Science and Technology, Wuhan University of Science and Technology, Wuhan, 430065, People's Republic of China
| | - Chen Zhang
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People's Republic of China
| |
Collapse
|
11
|
Cao J, Pickup S, Rosen M, Zhou R. Impact of Arterial Input Function and Pharmacokinetic Models on DCE-MRI Biomarkers for Detection of Vascular Effect Induced by Stroma-Directed Drug in an Orthotopic Mouse Model of Pancreatic Cancer. Mol Imaging Biol 2023:10.1007/s11307-023-01824-7. [PMID: 37166575 DOI: 10.1007/s11307-023-01824-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 04/28/2023] [Accepted: 05/01/2023] [Indexed: 05/12/2023]
Abstract
PURPOSE We demonstrated earlier in mouse models of pancreatic ductal adenocarcinoma (PDA) that Ktrans derived from dynamic contrast-enhanced (DCE) MRI detected microvascular effect induced by PEGPH20, a hyaluronidase which removes stromal hyaluronan, leading to reduced interstitial fluid pressure in the tumor (Clinical Cancer Res (2019) 25: 2314-2322). How the choice of pharmacokinetic (PK) model and arterial input function (AIF) may impact DCE-derived markers for detecting such an effect is not known. PROCEDURES Retrospective analyses of the DCE-MRI of the orthotopic PDA model are performed to examine the impact of individual versus group AIF combined with Tofts model (TM), extended-Tofts model (ETM), or shutter-speed model (SSM) on the ability to detect the microvascular changes induced by PEGPH20 treatment. RESULTS Individual AIF exhibit a marked difference in peak gadolinium concentration. However, across all three PK models, kep values show a significant correlation between individual versus group-AIF (p < 0.01). Regardless individual or group AIF, when kep is obtained from fitting the DCE-MRI data using the SSM, kep shows a significant increase after PEGPH20 treatment (p < 0.05 compared to the baseline); %change of kep from baseline to post-treatment is also significantly different between PEGPH20 versus vehicle group (p < 0.05). In comparison, when kep is derived from the TM, only the use of individual AIF leads to a significant increase of kep after PEGPH20 treatment, whereas the %change of kep is not different between PEGPH20 versus vehicle group. Group AIF but not individual AIF allows detection of a significant increase of Vp (derived from the ETM) in PEGPH20 versus vehicle group (p < 0.05). Increase of Vp is consistent with a large increase of mean capillary lumen area estimated from immunostaining. CONCLUSION Our results suggest that kep derived from SSM and Vp from ETM, both using group AIF, are optimal for the detection of microvascular changes induced by stroma-directed drug PEGPH20. These analyses provide insights in the choice of PK model and AIF for optimal DCE protocol design in mouse pancreatic cancer models.
Collapse
Affiliation(s)
- Jianbo Cao
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Current address: Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
| | - Stephen Pickup
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Mark Rosen
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Pancreatic Cancer Research Center, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Rong Zhou
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Pancreatic Cancer Research Center, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
12
|
Rastogi A, Dutta A, Yalavarthy PK. VTDCE-Net: A time invariant deep neural network for direct estimation of pharmacokinetic parameters from undersampled DCE MRI data. Med Phys 2023; 50:1560-1572. [PMID: 36354289 DOI: 10.1002/mp.16081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 10/17/2022] [Accepted: 10/19/2022] [Indexed: 11/12/2022] Open
Abstract
PURPOSE To propose a robust time and space invariant deep learning (DL) method to directly estimate the pharmacokinetic/tracer kinetic (PK/TK) parameters from undersampled dynamic contrast-enhanced (DCE) magnetic resonance imaging (MRI) data. METHODS DCE-MRI consists of 4D (3D-spatial + temporal) data and has been utilized to estimate 3D (spatial) tracer kinetic maps. Existing DL architecture for this task needs retraining for variation in temporal and/or spatial dimensions. This work proposes a DL algorithm that is invariant to training and testing in both temporal and spatial dimensions. The proposed network was based on a 2.5-dimensional Unet architecture, where the encoder consists of a 3D convolutional layer and the decoder consists of a 2D convolutional layer. The proposed VTDCE-Net was evaluated for solving the ill-posed inverse problem of directly estimating TK parameters from undersampled k - t $k-t$ space data of breast cancer patients, and the results were systematically compared with a total variation (TV) regularization based direct parameter estimation scheme. In the breast dataset, the training was performed on patients with 32 time samples, and testing was carried out on patients with 26 and 32 time samples. Translation of the proposed VTDCE-Net for brain dataset to show the generalizability was also carried out. Undersampling rates (R) of 8× , 12× , and 20× were utilized with PSNR and SSIM as the figures of merit in this evaluation. TK parameter maps estimated from fully sampled data were utilized as ground truth. RESULTS Experiments carried out in this work demonstrate that the proposed VTDCE-Net outperforms the TV scheme on both breast and brain datasets across all undersampling rates. For K trans $\mathbf {K_{trans}}$ and V p $\mathbf {V_{p}}$ maps, the improvement over TV is as high as 2 and 5 dB, respectively, using the proposed VTDCE-Net. CONCLUSION Temporal points invariant DL network that was proposed in this work to estimate the TK-parameters using DCE-MRI data has provided state-of-the-art performance compared to standard image reconstruction methods and is shown to work across all undersampling rates.
Collapse
Affiliation(s)
- Aditya Rastogi
- Computational and Data Sciences, Indian Institute of Science, Bengaluru, 560012, India
| | - Arindam Dutta
- Computational and Data Sciences, Indian Institute of Science, Bengaluru, 560012, India
| | | |
Collapse
|
13
|
KALA D, ŠULC V, OLŠEROVÁ A, SVOBODA J, PRYSIAZHNIUK Y, POŠUSTA A, KYNČL M, ŠANDA J, TOMEK A, OTÁHAL J. Evaluation of blood-brain barrier integrity by the analysis of dynamic contrast-enhanced MRI - a comparison of quantitative and semi-quantitative methods. Physiol Res 2022; 71:S259-S275. [PMID: 36647914 PMCID: PMC9906669 DOI: 10.33549/physiolres.934998] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Disruption of the blood-brain barrier (BBB) is a key feature of various brain disorders. To assess its integrity a parametrization of dynamic magnetic resonance imaging (DCE MRI) with a contrast agent (CA) is broadly used. Parametrization can be done quantitatively or semi-quantitatively. Quantitative methods directly describe BBB permeability but exhibit several drawbacks such as high computation demands, reproducibility issues, or low robustness. Semi-quantitative methods are fast to compute, simply mathematically described, and robust, however, they do not describe the status of BBB directly but only as a variation of CA concentration in measured tissue. Our goal was to elucidate differences between five semi-quantitative parameters: maximal intensity (Imax), normalized permeability index (NPI), and difference in DCE values between three timepoints: baseline, 5 min, and 15 min (delta5-0, delta15-0, delta15-5) and two quantitative parameters: transfer constant (Ktrans) and an extravascular fraction (Ve). For the purpose of comparison, we analyzed DCE data of four patients 12-15 days after the stroke with visible CA enhancement. Calculated parameters showed abnormalities spatially corresponding with the ischemic lesion, however, findings in individual parameters morphometrically differed. Ktrans and Ve were highly correlated. Delta5-0 and delta15-0 were prominent in regions with rapid CA enhancement and highly correlated with Ktrans. Abnormalities in delta15-5 and NPI were more homogenous with less variable values, smoother borders, and less detail than Ktrans. Moreover, only delta15-5 and NPI were able to distinguish vessels from extravascular space. Our comparison provides important knowledge for understanding and interpreting parameters derived from DCE MRI by both quantitative and semi-quantitative methods.
Collapse
Affiliation(s)
- David KALA
- Laboratory of Developmental Epileptology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic,Faculty of Electrical Engineering, Czech Technical University in Prague, Czech Republic
| | - Vlastimil ŠULC
- Department of Neurology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Anna OLŠEROVÁ
- Department of Neurology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Jan SVOBODA
- Laboratory of Developmental Epileptology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Yeva PRYSIAZHNIUK
- Laboratory of Developmental Epileptology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Antonín POŠUSTA
- Laboratory of Developmental Epileptology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Martin KYNČL
- Department of Radiology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Jan ŠANDA
- Department of Radiology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Aleš TOMEK
- Department of Neurology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Jakub OTÁHAL
- Laboratory of Developmental Epileptology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic,Department of Pathophysiology, Second Faculty of Medicine, Charles University, Czech Republic
| |
Collapse
|
14
|
Ye S, Lim JY, Huang W. Statistical considerations for repeatability and reproducibility of quantitative imaging biomarkers. BJR Open 2022; 4:20210083. [PMID: 36452056 PMCID: PMC9667479 DOI: 10.1259/bjro.20210083] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 07/25/2022] [Accepted: 07/26/2022] [Indexed: 11/05/2022] Open
Abstract
Quantitative imaging biomarkers (QIBs) are increasingly used in clinical studies. Because many QIBs are derived through multiple steps in image data acquisition and data analysis, QIB measurements can produce large variabilities, posing a significant challenge in translating QIBs into clinical trials, and ultimately, clinical practice. Both repeatability and reproducibility constitute the reliability of a QIB measurement. In this article, we review the statistical aspects of repeatability and reproducibility of QIB measurements by introducing methods and metrics for assessments of QIB repeatability and reproducibility and illustrating the impact of QIB measurement error on sample size and statistical power calculations, as well as predictive performance with a QIB as a predictive biomarker.
Collapse
Affiliation(s)
- Shangyuan Ye
- Biostatistics Shared Resource, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, United States
| | - Jeong Youn Lim
- Biostatistics Shared Resource, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, United States
| | - Wei Huang
- Advanced Imaging Research Center, Oregon Health and Science University, Portland, OR, United States
| |
Collapse
|
15
|
Amini Farsani Z, Schmid VJ. Maximum Entropy Technique and Regularization Functional for Determining the Pharmacokinetic Parameters in DCE-MRI. J Digit Imaging 2022; 35:1176-1188. [PMID: 35618849 PMCID: PMC9582183 DOI: 10.1007/s10278-022-00646-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 04/15/2022] [Accepted: 04/21/2022] [Indexed: 10/31/2022] Open
Abstract
This paper aims to solve the arterial input function (AIF) determination in dynamic contrast-enhanced MRI (DCE-MRI), an important linear ill-posed inverse problem, using the maximum entropy technique (MET) and regularization functionals. In addition, estimating the pharmacokinetic parameters from a DCE-MR image investigations is an urgent need to obtain the precise information about the AIF-the concentration of the contrast agent on the left ventricular blood pool measured over time. For this reason, the main idea is to show how to find a unique solution of linear system of equations generally in the form of [Formula: see text] named an ill-conditioned linear system of equations after discretization of the integral equations, which appear in different tomographic image restoration and reconstruction issues. Here, a new algorithm is described to estimate an appropriate probability distribution function for AIF according to the MET and regularization functionals for the contrast agent concentration when applying Bayesian estimation approach to estimate two different pharmacokinetic parameters. Moreover, by using the proposed approach when analyzing simulated and real datasets of the breast tumors according to pharmacokinetic factors, it indicates that using Bayesian inference-that infer the uncertainties of the computed solutions, and specific knowledge of the noise and errors-combined with the regularization functional of the maximum entropy problem, improved the convergence behavior and led to more consistent morphological and functional statistics and results. Finally, in comparison to the proposed exponential distribution based on MET and Newton's method, or Weibull distribution via the MET and teaching-learning-based optimization (MET/TLBO) in the previous studies, the family of Gamma and Erlang distributions estimated by the new algorithm are more appropriate and robust AIFs.
Collapse
Affiliation(s)
- Zahra Amini Farsani
- Bayesian Imaging and Spatial Statistics Group, Institute of Statistics, Ludwig-Maximilian-Universität München, Ludwigstraße 33, 80539, Munich, Germany. .,Statistics Department, School of Science, Lorestan University, 68151-44316, Khorramabad, Iran.
| | - Volker J Schmid
- Bayesian Imaging and Spatial Statistics Group, Institute of Statistics, Ludwig-Maximilian-Universität München, Ludwigstraße 33, 80539, Munich, Germany
| |
Collapse
|
16
|
Modified Maximum Entropy Method and Estimating the AIF via DCE-MRI Data Analysis. ENTROPY 2022; 24:e24020155. [PMID: 35205451 PMCID: PMC8871336 DOI: 10.3390/e24020155] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/16/2022] [Accepted: 01/17/2022] [Indexed: 02/06/2023]
Abstract
Background: For the kinetic models used in contrast-based medical imaging, the assignment of the arterial input function named AIF is essential for the estimation of the physiological parameters of the tissue via solving an optimization problem. Objective: In the current study, we estimate the AIF relayed on the modified maximum entropy method. The effectiveness of several numerical methods to determine kinetic parameters and the AIF is evaluated—in situations where enough information about the AIF is not available. The purpose of this study is to identify an appropriate method for estimating this function. Materials and Methods: The modified algorithm is a mixture of the maximum entropy approach with an optimization method, named the teaching-learning method. In here, we applied this algorithm in a Bayesian framework to estimate the kinetic parameters when specifying the unique form of the AIF by the maximum entropy method. We assessed the proficiency of the proposed method for assigning the kinetic parameters in the dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI), when determining AIF with some other parameter-estimation methods and a standard fixed AIF method. A previously analyzed dataset consisting of contrast agent concentrations in tissue and plasma was used. Results and Conclusions: We compared the accuracy of the results for the estimated parameters obtained from the MMEM with those of the empirical method, maximum likelihood method, moment matching (“method of moments”), the least-square method, the modified maximum likelihood approach, and our previous work. Since the current algorithm does not have the problem of starting point in the parameter estimation phase, it could find the best and nearest model to the empirical model of data, and therefore, the results indicated the Weibull distribution as an appropriate and robust AIF and also illustrated the power and effectiveness of the proposed method to estimate the kinetic parameters.
Collapse
|
17
|
Semi-automated and interactive segmentation of contrast-enhancing masses on breast DCE-MRI using spatial fuzzy clustering. Biomed Signal Process Control 2022. [DOI: 10.1016/j.bspc.2021.103113] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
18
|
Jarrett AM, Kazerouni AS, Wu C, Virostko J, Sorace AG, DiCarlo JC, Hormuth DA, Ekrut DA, Patt D, Goodgame B, Avery S, Yankeelov TE. Quantitative magnetic resonance imaging and tumor forecasting of breast cancer patients in the community setting. Nat Protoc 2021; 16:5309-5338. [PMID: 34552262 PMCID: PMC9753909 DOI: 10.1038/s41596-021-00617-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 08/12/2021] [Indexed: 02/07/2023]
Abstract
This protocol describes a complete data acquisition, analysis and computational forecasting pipeline for employing quantitative MRI data to predict the response of locally advanced breast cancer to neoadjuvant therapy in a community-based care setting. The methodology has previously been successfully applied to a heterogeneous patient population. The protocol details how to acquire the necessary images followed by registration, segmentation, quantitative perfusion and diffusion analysis, model calibration, and prediction. The data collection portion of the protocol requires ~25 min of scanning, postprocessing requires 2-3 h, and the model calibration and prediction components require ~10 h per patient depending on tumor size. The response of individual breast cancer patients to neoadjuvant therapy is forecast by application of a biophysical, reaction-diffusion mathematical model to these data. Successful application of the protocol results in coregistered MRI data from at least two scan visits that quantifies an individual tumor's size, cellularity and vascular properties. This enables a spatially resolved prediction of how a particular patient's tumor will respond to therapy. Expertise in image acquisition and analysis, as well as the numerical solution of partial differential equations, is required to carry out this protocol.
Collapse
Affiliation(s)
- Angela M Jarrett
- Oden Institute for Computational Engineering and Sciences, Austin, TX, USA
- Livestrong Cancer Institutes, Austin, TX, USA
| | - Anum S Kazerouni
- Departments of Biomedical Engineering, Austin, TX, USA
- Department of Radiology, University of Washington, Seattle, WA, USA
| | - Chengyue Wu
- Oden Institute for Computational Engineering and Sciences, Austin, TX, USA
| | - John Virostko
- Livestrong Cancer Institutes, Austin, TX, USA
- Departments of Diagnostic Medicine, Austin, TX, USA
- Departments of Oncology, Austin, TX, USA
| | - Anna G Sorace
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Julie C DiCarlo
- Oden Institute for Computational Engineering and Sciences, Austin, TX, USA
- Livestrong Cancer Institutes, Austin, TX, USA
| | - David A Hormuth
- Oden Institute for Computational Engineering and Sciences, Austin, TX, USA
- Livestrong Cancer Institutes, Austin, TX, USA
| | - David A Ekrut
- Oden Institute for Computational Engineering and Sciences, Austin, TX, USA
| | | | - Boone Goodgame
- Departments of Oncology, Austin, TX, USA
- Departments of Internal Medicine, The University of Texas at Austin, Austin, Texas, USA
- Seton Hospital, Austin, TX, USA
| | - Sarah Avery
- Austin Radiological Association, Austin, TX, USA
| | - Thomas E Yankeelov
- Oden Institute for Computational Engineering and Sciences, Austin, TX, USA.
- Livestrong Cancer Institutes, Austin, TX, USA.
- Departments of Biomedical Engineering, Austin, TX, USA.
- Departments of Diagnostic Medicine, Austin, TX, USA.
- Departments of Oncology, Austin, TX, USA.
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
19
|
Xing Y, Duan Y, P Indurkar P, Qiu A, Chen N. Optical breast atlas as a testbed for image reconstruction in optical mammography. Sci Data 2021; 8:257. [PMID: 34593824 PMCID: PMC8484607 DOI: 10.1038/s41597-021-01037-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 08/16/2021] [Indexed: 11/25/2022] Open
Abstract
We present two optical breast atlases for optical mammography, aiming to advance the image reconstruction research by providing a common platform to test advanced image reconstruction algorithms. Each atlas consists of five individual breast models. The first atlas provides breast vasculature surface models, which are derived from human breast dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) data using image segmentation. A finite element-based method is used to deform the breast vasculature models from their natural shapes to generate the second atlas, compressed breast models. Breast compression is typically done in X-ray mammography but also necessary for some optical mammography systems. Technical validation is presented to demonstrate how the atlases can be used to study the image reconstruction algorithms. Optical measurements are generated numerically with compressed breast models and a predefined configuration of light sources and photodetectors. The simulated data is fed into three standard image reconstruction algorithms to reconstruct optical images of the vasculature, which can then be compared with the ground truth to evaluate their performance.
Collapse
Affiliation(s)
- Yidan Xing
- Biomedical Engineering, National University of Singapore, Singapore, Singapore
| | - Yubo Duan
- Hangzhou One-North Medical Technologies, Hangzhou, China
| | - Padmeya P Indurkar
- Mechanical Engineering, National University of Singapore, Singapore, Singapore
| | - Anqi Qiu
- Biomedical Engineering, National University of Singapore, Singapore, Singapore
| | - Nanguang Chen
- Biomedical Engineering, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
20
|
Feasibility Study on Using Dynamic Contrast Enhanced MRI to Assess the Effect of Tyrosine Kinase Inhibitor Therapy within the STAR Trial of Metastatic Renal Cell Cancer. Diagnostics (Basel) 2021; 11:diagnostics11071302. [PMID: 34359384 PMCID: PMC8306403 DOI: 10.3390/diagnostics11071302] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 07/06/2021] [Accepted: 07/16/2021] [Indexed: 01/04/2023] Open
Abstract
Objective: To identify dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) parameters predictive of early disease progression in patients with metastatic renal cell cancer (mRCC) treated with anti-angiogenic tyrosine kinase inhibitors (TKI). Methods: The study was linked to a phase II/III randomised control trial. Patients underwent DCE-MRI before, at 4- and 10-weeks after initiation of TKI. DCE-MRI parameters at each time-point were derived from a single-compartment tracer kinetic model, following semi-automated tumour segmentation by two independent readers. Primary endpoint was correlation of DCE-MRI parameters with disease progression at 6-months. Receiver operating characteristic (ROC) curve analysis and area under the curve (AUC) values were calculated for parameters associated with disease progression at 6 months. Inter-observer agreement was assessed using the intraclass correlation coefficient (ICC). Results: 23 tumours in 14 patients were measurable. Three patients had disease progression at 6 months. The percentage (%) change in perfused tumour volume between baseline and 4-week DCE-MRI (p = 0.016), mean transfer constant Ktrans change (p = 0.038), and % change in extracellular volume (p = 0.009) between 4- and 10-week MRI, correlated with early disease progression (AUC 0.879 for each parameter). Inter-observer agreement was excellent for perfused tumour volume, Ktrans and extracellular volume (ICC: 0.928, 0.949, 0.910 respectively). Conclusions: Early measurement of DCE-MRI biomarkers of tumour perfusion at 4- and 10-weeks predicts disease progression at 6-months following TKI therapy in mRCC.
Collapse
|
21
|
Li K, Machireddy A, Tudorica A, Moloney B, Oh KY, Jafarian N, Partridge SC, Li X, Huang W. Discrimination of Malignant and Benign Breast Lesions Using Quantitative Multiparametric MRI: A Preliminary Study. ACTA ACUST UNITED AC 2021; 6:148-159. [PMID: 32548291 PMCID: PMC7289240 DOI: 10.18383/j.tom.2019.00028] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
We aimed to compare diagnostic performance in discriminating malignant and benign breast lesions between two intravoxel incoherent motion (IVIM) analysis methods for diffusion-weighted magnetic resonance imaging (DW-MRI) data and between DW- and dynamic contrast-enhanced (DCE)-MRI, and to determine if combining DW- and DCE-MRI further improves diagnostic accuracy. DW-MRI with 12 b-values and DCE-MRI were performed on 26 patients with 28 suspicious breast lesions before biopsies. The traditional biexponential fitting and a 3-b-value method were used for independent IVIM analysis of the DW-MRI data. Simulations were performed to evaluate errors in IVIM parameter estimations by the two methods across a range of signal-to-noise ratio (SNR). Pharmacokinetic modeling of DCE-MRI data was performed. Conventional radiological MRI reading yielded 86% sensitivity and 21% specificity in breast cancer diagnosis. At the same sensitivity, specificity of individual DCE- and DW-MRI markers improved to 36%–57% and that of combined DCE- or combined DW-MRI markers to 57%–71%, with DCE-MRI markers showing better diagnostic performance. The combination of DCE- and DW-MRI markers further improved specificity to 86%–93% and the improvements in diagnostic accuracy were statistically significant (P < .05) when compared with standard clinical MRI reading and most individual markers. At low breast DW-MRI SNR values (<50), like those typically seen in clinical studies, the 3-b-value approach for IVIM analysis generates markers with smaller errors and with comparable or better diagnostic performances compared with biexponential fitting. This suggests that the 3-b-value method could be an optimal IVIM-MRI method to be combined with DCE-MRI for improved diagnostic accuracy.
Collapse
Affiliation(s)
- Kurt Li
- International School of Beaverton, Aloha, OR
| | - Archana Machireddy
- Center for Spoken Language Understanding, Oregon Health & Science University, Portland, OR
| | - Alina Tudorica
- Department of Diagnostic Radiology, Oregon Health & Science University, Portland, OR
| | - Brendan Moloney
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, OR; and
| | - Karen Y Oh
- Department of Diagnostic Radiology, Oregon Health & Science University, Portland, OR
| | - Neda Jafarian
- Department of Diagnostic Radiology, Oregon Health & Science University, Portland, OR
| | | | - Xin Li
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, OR; and
| | - Wei Huang
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, OR; and
| |
Collapse
|
22
|
Characterizing Errors in Pharmacokinetic Parameters from Analyzing Quantitative Abbreviated DCE-MRI Data in Breast Cancer. ACTA ACUST UNITED AC 2021; 7:253-267. [PMID: 34201654 PMCID: PMC8293327 DOI: 10.3390/tomography7030023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 06/15/2021] [Accepted: 06/21/2021] [Indexed: 12/13/2022]
Abstract
This study characterizes the error that results when performing quantitative analysis of abbreviated dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) data of the breast with the Standard Kety-Tofts (SKT) model and its Patlak variant. More specifically, we used simulations and patient data to determine the accuracy with which abbreviated time course data could reproduce the pharmacokinetic parameters, Ktrans (volume transfer constant) and ve (extravascular/extracellular volume fraction), when compared to the full time course data. SKT analysis of simulated abbreviated time courses (ATCs) based on the imaging parameters from two available datasets (collected with a 3T MRI scanner) at a temporal resolution of 15 s (N = 15) and 7.23 s (N = 15) found a concordance correlation coefficient (CCC) greater than 0.80 for ATCs of length 3.0 and 2.5 min, respectively, for the Ktrans parameter. Analysis of the experimental data found that at least 90% of patients met this CCC cut-off of 0.80 for the ATCs of the aforementioned lengths. Patlak analysis of experimental data found that 80% of patients from the 15 s resolution dataset and 90% of patients from the 7.27 s resolution dataset met the 0.80 CCC cut-off for ATC lengths of 1.25 and 1.09 min, respectively. This study provides evidence for both the feasibility and potential utility of performing a quantitative analysis of abbreviated breast DCE-MRI in conjunction with acquisition of current standard-of-care high resolution scans without significant loss of information in the community setting.
Collapse
|
23
|
McGee KP, Hwang KP, Sullivan DC, Kurhanewicz J, Hu Y, Wang J, Li W, Debbins J, Paulson E, Olsen JR, Hua CH, Warner L, Ma D, Moros E, Tyagi N, Chung C. Magnetic resonance biomarkers in radiation oncology: The report of AAPM Task Group 294. Med Phys 2021; 48:e697-e732. [PMID: 33864283 PMCID: PMC8361924 DOI: 10.1002/mp.14884] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 03/24/2021] [Accepted: 03/28/2021] [Indexed: 12/16/2022] Open
Abstract
A magnetic resonance (MR) biologic marker (biomarker) is a measurable quantitative characteristic that is an indicator of normal biological and pathogenetic processes or a response to therapeutic intervention derived from the MR imaging process. There is significant potential for MR biomarkers to facilitate personalized approaches to cancer care through more precise disease targeting by quantifying normal versus pathologic tissue function as well as toxicity to both radiation and chemotherapy. Both of which have the potential to increase the therapeutic ratio and provide earlier, more accurate monitoring of treatment response. The ongoing integration of MR into routine clinical radiation therapy (RT) planning and the development of MR guided radiation therapy systems is providing new opportunities for MR biomarkers to personalize and improve clinical outcomes. Their appropriate use, however, must be based on knowledge of the physical origin of the biomarker signal, the relationship to the underlying biological processes, and their strengths and limitations. The purpose of this report is to provide an educational resource describing MR biomarkers, the techniques used to quantify them, their strengths and weakness within the context of their application to radiation oncology so as to ensure their appropriate use and application within this field.
Collapse
Affiliation(s)
- Kiaran P McGee
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Ken-Pin Hwang
- Department of Imaging Physics, Division of Diagnostic Imaging, MD Anderson Cancer Center, University of Texas, Houston, Texas, USA
| | - Daniel C Sullivan
- Department of Radiology, Duke University, Durham, North Carolina, USA
| | - John Kurhanewicz
- Department of Radiology, University of California, San Francisco, California, USA
| | - Yanle Hu
- Department of Radiation Oncology, Mayo Clinic, Scottsdale, Arizona, USA
| | - Jihong Wang
- Department of Radiation Oncology, MD Anderson Cancer Center, University of Texas, Houston, Texas, USA
| | - Wen Li
- Department of Radiation Oncology, University of Arizona, Tucson, Arizona, USA
| | - Josef Debbins
- Department of Radiology, Barrow Neurologic Institute, Phoenix, Arizona, USA
| | - Eric Paulson
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Jeffrey R Olsen
- Department of Radiation Oncology, University of Colorado Denver - Anschutz Medical Campus, Denver, Colorado, USA
| | - Chia-Ho Hua
- Department of Radiation Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | | | - Daniel Ma
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Eduardo Moros
- Department of Radiation Oncology, Moffitt Cancer Center, Tampa, Florida, USA
| | - Neelam Tyagi
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Caroline Chung
- Department of Radiation Oncology, MD Anderson Cancer Center, University of Texas, Houston, Texas, USA
| |
Collapse
|
24
|
Xie Y, Zhao J, Zhang P. A multicompartment model for intratumor tissue-specific analysis of DCE-MRI using non-negative matrix factorization. Med Phys 2021; 48:2400-2411. [PMID: 33608885 DOI: 10.1002/mp.14793] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 12/22/2020] [Accepted: 01/29/2021] [Indexed: 11/12/2022] Open
Abstract
PURPOSE A pharmacokinetic analysis of dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) data is subject to inaccuracy and instability partly owing to the partial volume effect (PVE). We proposed a new multicompartment model for a tissue-specific pharmacokinetic analysis in DCE-MRI data to solve the PVE problem and to provide better kinetic parameter maps. METHODS We introduced an independent parameter named fractional volumes of tissue compartments in each DCE-MRI pixel to construct a new linear separable multicompartment model, which simultaneously estimates the pixel-wise time-concentration curves and fractional volumes without the need of the pure-pixel assumption. This simplified convex optimization model was solved using a special type of non-negative matrix factorization (NMF) algorithm called the minimum-volume constraint NMF (MVC-NMF). RESULTS To test the model, synthetic datasets were established based on the general pharmacokinetic parameters. On well-designed synthetic data, the proposed model reached lower bias and lower root mean square fitting error compared to the state-of-the-art algorithm in different noise levels. In addition, the real dataset from QIN-BREAST-DCE-MRI was analyzed, and we observed an improved pharmacokinetic parameter estimation to distinguish the treatment response to chemotherapy applied to breast cancer. CONCLUSION Our model improved the accuracy and stability of the tissue-specific estimation of the fractional volumes and kinetic parameters in DCE-MRI data, and improved the robustness to noise, providing more accurate kinetics for more precise prognosis and therapeutic response evaluation using DCE-MRI.
Collapse
Affiliation(s)
- Yuhai Xie
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Jun Zhao
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Puming Zhang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| |
Collapse
|
25
|
Rastogi A, Yalavarthy PK. SpiNet: A deep neural network for Schatten p-norm regularized medical image reconstruction. Med Phys 2021; 48:2214-2229. [PMID: 33525049 DOI: 10.1002/mp.14744] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 12/30/2020] [Accepted: 01/19/2021] [Indexed: 11/07/2022] Open
Abstract
PURPOSE To propose a generic deep learning based medical image reconstruction model (named as SpiNet) that can enforce any Schatten p-norm regularization with 0 < p ≤ 2, where the p can be learnt (or fixed) based on the problem at hand. METHODS Model-based deep learning architecture for solving inverse problems consists of two parts, a deep learning based denoiser and an iterative data consistency solver. The former has either L2 norm or L1 norm enforced on it, which are convex and can be easily minimized. This work proposes a method to enforce any p norm on the noise prior where 0 < p ≤ 2. This is achieved by using Majorization-Minimization algorithm, which upper bounds the cost function with a convex function, thus can be easily minimized. The proposed SpiNet has the capability to work for a fixed p or it can learn p based on the data. The network was tested for solving the inverse problem of reconstructing magnetic resonance (MR) images from undersampled k space data and the results were compared with a popular model-based deep learning architecture MoDL which enforces L2 norm along with other compressive sensing-based algorithms. This comparison between MoDL and proposed SpiNet was performed for undersampling rates (R) of 2×, 4×, 6×, 8×, 12×, 16×, and 20×. Multiple figures of merit such as PSNR, SSIM, and NRMSE were utilized in this comparison. A two-tailed t test was performed for all undersampling rates and for all metrices for proving the superior performance of proposed SpiNet compared to MoDL. For training and testing, the same dataset that was utilized in MoDL implementation was deployed. RESULTS The results indicate that for all undersampling rates, the proposed SpiNet shows higher PSNR and SSIM and lower NRMSE than MoDL. However, for low undersampling rates of 2× and 4×, there is no significant difference in performance of proposed SpiNet and MoDL in terms of PSNR and NRMSE. This can be expected as the learnt p value is close to 2 (norm enforced by MoDL). For higher undersampling rates ≥6×, SpiNet significantly outperforms MoDL in all metrices with improvement as high as 4 dB in PSNR and 0.5 points in SSIM. CONCLUSION As deep learning based medical image reconstruction methods are gaining popularity, the proposed SpiNet provides a generic framework to incorporate Schatten p-norm regularization with 0 <p ≤ 2 with an added advantage of providing superior performance compared to its counterparts. The proposed SpiNet also has useful addition of Schatten p-norm value in regularization term being automatically chosen based on the available training data.
Collapse
Affiliation(s)
- Aditya Rastogi
- Department of Computational and Data Sciences, Indian Institute of Science, Bangalore, 560012, India
| | | |
Collapse
|
26
|
Hesse LS, Kuling G, Veta M, Martel AL. Intensity Augmentation to Improve Generalizability of Breast Segmentation Across Different MRI Scan Protocols. IEEE Trans Biomed Eng 2021; 68:759-770. [DOI: 10.1109/tbme.2020.3016602] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
27
|
Rastogi A, Yalavarthy PK. Comparison of iterative parametric and indirect deep learning‐based reconstruction methods in highly undersampled DCE‐MR Imaging of the breast. Med Phys 2020; 47:4838-4861. [DOI: 10.1002/mp.14447] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 07/24/2020] [Accepted: 08/03/2020] [Indexed: 12/23/2022] Open
Affiliation(s)
- Aditya Rastogi
- Department of Computational and Data Sciences Indian Institute of Science Bangalore560012 India
| | | |
Collapse
|
28
|
Optimal Control Theory for Personalized Therapeutic Regimens in Oncology: Background, History, Challenges, and Opportunities. J Clin Med 2020; 9:jcm9051314. [PMID: 32370195 PMCID: PMC7290915 DOI: 10.3390/jcm9051314] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 04/25/2020] [Accepted: 04/28/2020] [Indexed: 12/13/2022] Open
Abstract
Optimal control theory is branch of mathematics that aims to optimize a solution to a dynamical system. While the concept of using optimal control theory to improve treatment regimens in oncology is not novel, many of the early applications of this mathematical technique were not designed to work with routinely available data or produce results that can eventually be translated to the clinical setting. The purpose of this review is to discuss clinically relevant considerations for formulating and solving optimal control problems for treating cancer patients. Our review focuses on two of the most widely used cancer treatments, radiation therapy and systemic therapy, as they naturally lend themselves to optimal control theory as a means to personalize therapeutic plans in a rigorous fashion. To provide context for optimal control theory to address either of these two modalities, we first discuss the major limitations and difficulties oncologists face when considering alternate regimens for their patients. We then provide a brief introduction to optimal control theory before formulating the optimal control problem in the context of radiation and systemic therapy. We also summarize examples from the literature that illustrate these concepts. Finally, we present both challenges and opportunities for dramatically improving patient outcomes via the integration of clinically relevant, patient-specific, mathematical models and optimal control theory.
Collapse
|
29
|
Bliesener Y, Acharya J, Nayak KS. Efficient DCE-MRI Parameter and Uncertainty Estimation Using a Neural Network. IEEE TRANSACTIONS ON MEDICAL IMAGING 2020; 39:1712-1723. [PMID: 31794389 PMCID: PMC8887912 DOI: 10.1109/tmi.2019.2953901] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Quantitative DCE-MRI provides voxel-wise estimates of tracer-kinetic parameters that are valuable in the assessment of health and disease. These maps suffer from many known sources of variability. This variability is expensive to compute using current methods, and is typically not reported. Here, we demonstrate a novel approach for simultaneous estimation of tracer-kinetic parameters and their uncertainty due to intrinsic characteristics of the tracer-kinetic model, with very low computation time. We train and use a neural network to estimate the approximate joint posterior distribution of tracer-kinetic parameters. Uncertainties are estimated for each voxel and are specific to the patient, exam, and lesion. We demonstrate the methods' ability to produce accurate tracer-kinetic maps. We compare predicted parameter ranges with uncertainties introduced by noise and by differences in post-processing in a digital reference object. The predicted parameter ranges correlate well with tracer-kinetic parameter ranges observed across different noise realizations and regression algorithms. We also demonstrate the value of this approach to differentiate significant from insignificant changes in brain tumor pharmacokinetics over time. This is achieved by enforcing consistency in resolving model singularities in the applied tracer-kinetic model.
Collapse
|
30
|
Fedorov A, Beichel R, Kalpathy-Cramer J, Clunie D, Onken M, Riesmeier J, Herz C, Bauer C, Beers A, Fillion-Robin JC, Lasso A, Pinter C, Pieper S, Nolden M, Maier-Hein K, Herrmann MD, Saltz J, Prior F, Fennessy F, Buatti J, Kikinis R. Quantitative Imaging Informatics for Cancer Research. JCO Clin Cancer Inform 2020; 4:444-453. [PMID: 32392097 PMCID: PMC7265794 DOI: 10.1200/cci.19.00165] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/16/2020] [Indexed: 01/06/2023] Open
Abstract
PURPOSE We summarize Quantitative Imaging Informatics for Cancer Research (QIICR; U24 CA180918), one of the first projects funded by the National Cancer Institute (NCI) Informatics Technology for Cancer Research program. METHODS QIICR was motivated by the 3 use cases from the NCI Quantitative Imaging Network. 3D Slicer was selected as the platform for implementation of open-source quantitative imaging (QI) tools. Digital Imaging and Communications in Medicine (DICOM) was chosen for standardization of QI analysis outputs. Support of improved integration with community repositories focused on The Cancer Imaging Archive (TCIA). Priorities included improved capabilities of the standard, toolkits and tools, reference datasets, collaborations, and training and outreach. RESULTS Fourteen new tools to support head and neck cancer, glioblastoma, and prostate cancer QI research were introduced and downloaded over 100,000 times. DICOM was amended, with over 40 correction proposals addressing QI needs. Reference implementations of the standard in a popular toolkit and standalone tools were introduced. Eight datasets exemplifying the application of the standard and tools were contributed. An open demonstration/connectathon was organized, attracting the participation of academic groups and commercial vendors. Integration of tools with TCIA was improved by implementing programmatic communication interface and by refining best practices for QI analysis results curation. CONCLUSION Tools, capabilities of the DICOM standard, and datasets we introduced found adoption and utility within the cancer imaging community. A collaborative approach is critical to addressing challenges in imaging informatics at the national and international levels. Numerous challenges remain in establishing and maintaining the infrastructure of analysis tools and standardized datasets for the imaging community. Ideas and technology developed by the QIICR project are contributing to the NCI Imaging Data Commons currently being developed.
Collapse
Affiliation(s)
- Andrey Fedorov
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | | | | | | | | | | | - Christian Herz
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | | | | | | | | | | | | | - Marco Nolden
- German Cancer Research Center, Heidelberg, Germany
| | | | - Markus D. Herrmann
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | | | - Fred Prior
- University of Arkansas for Medical Sciences, Little Rock, AR
| | - Fiona Fennessy
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | | | - Ron Kikinis
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
31
|
Fusco R, Granata V, Maio F, Sansone M, Petrillo A. Textural radiomic features and time-intensity curve data analysis by dynamic contrast-enhanced MRI for early prediction of breast cancer therapy response: preliminary data. Eur Radiol Exp 2020; 4:8. [PMID: 32026095 PMCID: PMC7002809 DOI: 10.1186/s41747-019-0141-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 12/05/2019] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND To investigate the potential of semiquantitative time-intensity curve parameters compared to textural radiomic features on arterial phase images by dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) for early prediction of breast cancer neoadjuvant therapy response. METHODS A retrospective study of 45 patients subjected to DCE-MRI by public datasets containing examination performed prior to the start of treatment and after the treatment first cycle ('QIN Breast DCE-MRI' and 'QIN-Breast') was performed. In total, 11 semiquantitative parameters and 50 texture features were extracted. Non-parametric test, receiver operating characteristic analysis with area under the curve (ROC-AUC), Spearman correlation coefficient, and Kruskal-Wallis test with Bonferroni correction were applied. RESULTS Fifteen patients with pathological complete response (pCR) and 30 patients with non-pCR were analysed. Significant differences in median values between pCR patients and non-pCR patients were found for entropy, long-run emphasis, and busyness among the textural features, for maximum signal difference, washout slope, washin slope, and standardised index of shape among the dynamic semiquantitative parameters. The standardised index of shape had the best results with a ROC-AUC of 0.93 to differentiate pCR versus non-pCR patients. CONCLUSIONS The standardised index of shape could become a clinical tool to differentiate, in the early stages of treatment, responding to non-responding patients.
Collapse
Affiliation(s)
- Roberta Fusco
- Radiology Division, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Via Mariano Semmola, Naples, Italy.
| | - Vincenza Granata
- Radiology Division, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Via Mariano Semmola, Naples, Italy
| | - Francesca Maio
- Radiology Division, Universita' Degli Stui di Napoli Federico II, Via Pansini, Naples, Italy
| | - Mario Sansone
- Department of Electrical Engineering and Information Technologies (DIETI), University of Naples Federico II, Via Claudio, Naples, Italy
| | - Antonella Petrillo
- Radiology Division, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Via Mariano Semmola, Naples, Italy
| |
Collapse
|
32
|
Aryal MP, Lee C, Hawkins PG, Chapman C, Eisbruch A, Mierzwa M, Cao Y. Real-Time Quantitative Assessment of Accuracy and Precision of Blood Volume Derived from DCE-MRI in Individual Patients During a Clinical Trial. ACTA ACUST UNITED AC 2020; 5:61-67. [PMID: 30854443 PMCID: PMC6403042 DOI: 10.18383/j.tom.2018.00029] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Accuracy and precision of quantitative imaging (QI) metrics should be assessed in real time in each patient during a clinical trial to support QI-based decision-making. We developed a framework for real-time quantitative assessment of QI metrics and evaluated accuracy and precision of dynamic contrast-enhanced (DCE)-magnetic resonance imaging (MRI)–derived blood volume (BV) in a clinical trial for head and neck cancers. Patients underwent DCE-MRI before and after 2 weeks of radiation therapy (2wkRT). A mean as a reference value and a repeatability coefficient (RC) of BV values established from n patients in cerebellum volumes of interest (VOIs), which were normal and affected little by therapy, served as accuracy and precision measurements. The BV maps of a new patient were called accurate and precise if the values in cerebellum VOIs and the difference between the 2 scans agreed with the respective mean and RC with 95% confidence. The new data could be used to update reference values. Otherwise, the data were flagged for further evaluation before use in the trial. BV maps from 62 patients enrolled on the trial were evaluated. Mean BV values were 2.21 (±0.14) mL/100 g pre-RT and 2.22 (±0.17) mL/100 g at 2wkRT; relative RC was 15.9%. The BV maps from 3 patients were identified to be inaccurate and imprecise before use in the clinical trial. Our framework of real-time quantitative assessment of QI metrics during a clinical trial can be translated to different QI metrics and organ-sites for supporting QI-based decision-making that warrants success of a clinical trial.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yue Cao
- Departments of Radiation Oncology.,Radiology; and.,Biomedical Engineering, University of Michigan, Ann Arbor, MI
| |
Collapse
|
33
|
Parra NA, Lu H, Choi J, Gage K, Pow-Sang J, Gillies RJ, Balagurunathan Y. Habitats in DCE-MRI to Predict Clinically Significant Prostate Cancers. ACTA ACUST UNITED AC 2020; 5:68-76. [PMID: 30854444 PMCID: PMC6403034 DOI: 10.18383/j.tom.2018.00037] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Prostate cancer identification and assessment of clinical significance continues to be a challenge. Routine multiparametric magnetic resonance imaging has shown to be useful in assessing disease progression. Although dynamic contrast-enhanced imaging (DCE) has the ability to characterize perfusion across time and has shown enormous utility, radiological assessment (Prostate Imaging-Reporting and Data System or PIRADS version 2) has limited its use owing to lack of consistency and nonquantitative nature. In our work, we propose a systematic methodology to quantify perfusion dynamics for the DCE imaging. Using these metrics, 7 different subregions or perfusion habitats of the targeted lesions are localized and related to clinical significance. We found that quantitative features describing the habitat based on the late area under the DCE time-activity curve was a good predictor of clinical significance disease. The best predictive feature in the habitat had an AUC of 0.82, CI [0.81–0.83].
Collapse
Affiliation(s)
| | - Hong Lu
- Departments of Cancer Physiology.,Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | | | | | | | | | | |
Collapse
|
34
|
Abstract
The Quantitative Imaging Network of the National Cancer Institute is in its 10th year of operation, and research teams within the network are developing and validating clinical decision support software tools to measure or predict the response of cancers to various therapies. As projects progress from development activities to validation of quantitative imaging tools and methods, it is important to evaluate the performance and clinical readiness of the tools before committing them to prospective clinical trials. A variety of tests, including special challenges and tool benchmarking, have been instituted within the network to prepare the quantitative imaging tools for service in clinical trials. This article highlights the benchmarking process and provides a current evaluation of several tools in their transition from development to validation.
Collapse
Affiliation(s)
- Keyvan Farahani
- Cancer Imaging Program, National Cancer Institute of NIH, Bethesda, MD
| | - Darrell Tata
- Cancer Imaging Program, National Cancer Institute of NIH, Bethesda, MD
| | | |
Collapse
|
35
|
Machireddy A, Thibault G, Tudorica A, Afzal A, Mishal M, Kemmer K, Naik A, Troxell M, Goranson E, Oh K, Roy N, Jafarian N, Holtorf M, Huang W, Song X. Early Prediction of Breast Cancer Therapy Response using Multiresolution Fractal Analysis of DCE-MRI Parametric Maps. ACTA ACUST UNITED AC 2020; 5:90-98. [PMID: 30854446 PMCID: PMC6403033 DOI: 10.18383/j.tom.2018.00046] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
We aimed to determine whether multiresolution fractal analysis of voxel-based dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) parametric maps can provide early prediction of breast cancer response to neoadjuvant chemotherapy (NACT). In total, 55 patients underwent 4 DCE-MRI examinations before, during, and after NACT. The shutter-speed model was used to analyze the DCE-MRI data and generate parametric maps within the tumor region of interest. The proposed multiresolution fractal method and the more conventional methods of single-resolution fractal, gray-level co-occurrence matrix, and run-length matrix were used to extract features from the parametric maps. Only the data obtained before and after the first NACT cycle were used to evaluate early prediction of response. With a training (N = 40) and testing (N = 15) data set, support vector machine was used to assess the predictive abilities of the features in classification of pathologic complete response versus non-pathologic complete response. Generally the multiresolution fractal features from individual maps and the concatenated features from all parametric maps showed better predictive performances than conventional features, with receiver operating curve area under the curve (AUC) values of 0.91 (all parameters) and 0.80 (Ktrans), in the training and testing sets, respectively. The differences in AUC were statistically significant (P < .05) for several parametric maps. Thus, multiresolution analysis that decomposes the texture at various spatial-frequency scales may more accurately capture changes in tumor vascular heterogeneity as measured by DCE-MRI, and therefore provide better early prediction of NACT response.
Collapse
Affiliation(s)
| | | | | | - Aneela Afzal
- Oregon Health and Science University, Portland, OR
| | - May Mishal
- Oregon Health and Science University, Portland, OR
| | | | - Arpana Naik
- Oregon Health and Science University, Portland, OR
| | | | | | - Karen Oh
- Oregon Health and Science University, Portland, OR
| | - Nicole Roy
- Oregon Health and Science University, Portland, OR
| | | | | | - Wei Huang
- Oregon Health and Science University, Portland, OR
| | - Xubo Song
- Oregon Health and Science University, Portland, OR
| |
Collapse
|
36
|
Shao J, Zhang Z, Liu H, Song Y, Yan Z, Wang X, Hou Z. DCE-MRI pharmacokinetic parameter maps for cervical carcinoma prediction. Comput Biol Med 2020; 118:103634. [PMID: 32174312 DOI: 10.1016/j.compbiomed.2020.103634] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 01/15/2020] [Accepted: 01/27/2020] [Indexed: 12/30/2022]
Abstract
Pharmacokinetic parameters estimated from dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) time course data enable the physio-biological interpretation of tissue angiogenesis. This study aims to develop machine learning approaches for cervical carcinoma prediction based on pharmacokinetic parameters. The performance of individual parameters was assessed in terms of their efficacy in differentiating cancerous tissue from normal cervix tissue. The effect of combining parameters was evaluated using the following two approaches: the first approach was based on support vector machines (SVMs) to combine the parameters from one pharmacokinetic model or across several models; the second approach was based on a novel method called APITL (artificial pharmacokinetic images for transfer learning), which was designed to fully utilize the comprehensive pharmacokinetic information acquired from DCE-MRI data. A "winner-takes-all" strategy was employed to consolidate the slice-wise prediction into subject-wise prediction. Experiments were carried out with a dataset comprising 36 patients with cervical cancer and 17 healthy subjects. The results demonstrated that parameter Ve, representing volume fraction of the extracellular extravascular space (EES), attained high discriminative power regardless of the pharmacokinetic model used for estimation. An approximately 10% improvement in the accuracy was achieved with the SVM approach. The APITL method further outperformed SVM and attained a subject-wise prediction accuracy of 94.3%. Our experiment demonstrated that APITL could predict cervical carcinoma with high accuracy and had potential in clinical applications.
Collapse
Affiliation(s)
| | - Zhuo Zhang
- Institute for Infocomm Research, Singapore.
| | | | - Ying Song
- Institute for Infocomm Research, Singapore
| | - Zhihan Yan
- The Second Affiliated Hospital of Wenzhou Medical University, China
| | - Xue Wang
- The Second Affiliated Hospital of Wenzhou Medical University, China
| | - Zujun Hou
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, China
| |
Collapse
|
37
|
Taylor E, Zhou J, Lindsay P, Foltz W, Cheung M, Siddiqui I, Hosni A, Amir AE, Kim J, Hill RP, Jaffray DA, Hedley DW. Quantifying Reoxygenation in Pancreatic Cancer During Stereotactic Body Radiotherapy. Sci Rep 2020; 10:1638. [PMID: 32005829 PMCID: PMC6994660 DOI: 10.1038/s41598-019-57364-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 12/18/2019] [Indexed: 02/05/2023] Open
Abstract
Hypoxia, the state of low oxygenation that often arises in solid tumours due to their high metabolism and irregular vasculature, is a major contributor to the resistance of tumours to radiation therapy (RT) and other treatments. Conventional RT extends treatment over several weeks or more, and nominally allows time for oxygen levels to increase ("reoxygenation") as cancer cells are killed by RT, mitigating the impact of hypoxia. Recent advances in RT have led to an increase in the use stereotactic body radiotherapy (SBRT), which delivers high doses in five or fewer fractions. For cancers such as pancreatic adenocarcinoma for which hypoxia varies significantly between patients, SBRT might not be optimal, depending on the extent to which reoxygenation occurs during its short duration. We used fluoro-5-deoxy-α-D-arabinofuranosyl)-2-nitroimidazole positron-emission tomography (FAZA-PET) imaging to quantify hypoxia before and after 5-fraction SBRT delivered to patient-derived pancreatic cancer xenografts orthotopically implanted in mice. An imaging technique using only the pre-treatment FAZA-PET scan and repeat dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) scans throughout treatment was able to predict the change in hypoxia. Our results support the further testing of this technique for imaging of reoxygenation in the clinic.
Collapse
Affiliation(s)
- Edward Taylor
- Radiation Medicine Program, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, Ontario, M5G 2M9, Canada
| | - Jitao Zhou
- Department of Abdominal Oncology, Cancer Center and Laboratory of Signal Transduction and Molecular Targeting Therapy, West China Hospital, Sichuan University, Chengdu, China
| | - Patricia Lindsay
- Radiation Medicine Program, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, Ontario, M5G 2M9, Canada
| | - Warren Foltz
- Radiation Medicine Program, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, Ontario, M5G 2M9, Canada
| | - May Cheung
- Ontario Cancer Institute, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, Ontario, M5G 2M9, Canada
| | - Iram Siddiqui
- Department of Pathology, Hospital for Sick Children, 555 University Avenue, Toronto, Ontario, M5G 1X8, Canada
| | - Ali Hosni
- Radiation Medicine Program, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, Ontario, M5G 2M9, Canada
| | - Ahmed El Amir
- Radiation Medicine Program, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, Ontario, M5G 2M9, Canada
| | - John Kim
- Radiation Medicine Program, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, Ontario, M5G 2M9, Canada
| | - Richard P Hill
- Radiation Medicine Program, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, Ontario, M5G 2M9, Canada
- Ontario Cancer Institute, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, Ontario, M5G 2M9, Canada
| | - David A Jaffray
- Radiation Medicine Program, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, Ontario, M5G 2M9, Canada
| | - David W Hedley
- Ontario Cancer Institute, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, Ontario, M5G 2M9, Canada.
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, Ontario, M5G 2M9, Canada.
| |
Collapse
|
38
|
Moustafa AFI, Kamal RM, Gomaa MMM, Mostafa S, Mubarak R, El-Adawy M. Quantitative mathematical objective evaluation of contrast-enhanced spectral mammogram in the assessment of response to neoadjuvant chemotherapy and prediction of residual disease in breast cancer. THE EGYPTIAN JOURNAL OF RADIOLOGY AND NUCLEAR MEDICINE 2019. [DOI: 10.1186/s43055-019-0041-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
The aim of the study is to initiate a new quantitative mathematical objective tool for evaluation of response to neoadjuvant chemotherapy (NAC) and prediction of residual disease in breast cancer using contrast-enhanced spectral mammography (CESM). Forty-two breast cancer patients scheduled for receiving NAC were included. All patients underwent two CESM examinations: pre and post NAC. To assess the response to neoadjuvant chemotherapy, we used a mathematical image analysis software that can calculate the difference in the intensity of enhancement between the pre and post neoadjuvant contrast images (MATLAB and Simulink) (Release 2013b). The proposed technique used the pre and post neoadjuvant contrast images as inputs. The technique consists of three main steps: (1) preprocessing, (2) extracting the region of interest (ROI), and (3) assessment of the response to chemotherapy by measuring the percentage of change in the intensity of enhancement of malignant lesions in the pre and post neoadjuvant CESM studies using a quantitative mathematical technique. This technique depends on the analysis of number of pixels included within the ROI. We compared this technique with the currently used method of evaluation: RECIST 1.1 (response evaluation criteria in solid tumors 1.1) and using another combined response evaluation approach using both RECIST 1.1 in addition to a subjective visual evaluation. Results were then correlated with the postoperative pathology evaluation using Miller–Payne grades. For statistical evaluation, patients were classified into responders and non-responders in all evaluation methods.
Results
According to the Miller–Payne criteria, 39/42 (92.9%) of the participants were responders (Miller–Payne grades III, IV, and IV) and 3/42 (7.1%) were non-responders (Miller–Payne grades I and II). Using the proposed technique, 39/39 (100%) were responders in comparison to 38/39 patients (97.4%) using the combined criteria and 34/39 (87.2%) using the RECIST 1.1 evaluation. The calculated correlation coefficient of the proposed quantitative objective mathematical technique, RECIST 1.1 criteria, and the combined method was 0.89, 0.59, and 0.69 respectively. With classification of patients into responder and non-responders, the objective mathematical evaluation showed higher sensitivity, positive and negative predictive values, and overall accuracy (100%, 97.5%, 100%, and 85.7% respectively) compared to RECIST 1.1 evaluation (87.2%, 97.1%, 28.6%, and 54.8% respectively) and the combined response method (97.4%, 97.4%, 66.7%, and 85.7% respectively).
Conclusion
Quantitative mathematical objective evaluation using CESM images allows objective quantitative and accurate evaluation of the response of breast cancer to chemotherapy and is recommended as an alternative to the subjective techniques as a part of the pre-operative workup.
Collapse
|
39
|
Mittermeier A, Ertl-Wagner B, Ricke J, Dietrich O, Ingrisch M. Bayesian pharmacokinetic modeling of dynamic contrast-enhanced magnetic resonance imaging: validation and application. Phys Med Biol 2019; 64:18NT02. [PMID: 31404913 DOI: 10.1088/1361-6560/ab3a5a] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Tracer-kinetic analysis of dynamic contrast-enhanced magnetic resonance imaging data is commonly performed with the well-known Tofts model and nonlinear least squares (NLLS) regression. This approach yields point estimates of model parameters, uncertainty of these estimates can be assessed e.g. by an additional bootstrapping analysis. Here, we present a Bayesian probabilistic modeling approach for tracer-kinetic analysis with a Tofts model, which yields posterior probability distributions of perfusion parameters and therefore promises a robust and information-enriched alternative based on a framework of probability distributions. In this manuscript, we use the quantitative imaging biomarkers alliance (QIBA) Tofts phantom to evaluate the Bayesian tofts model (BTM) against a bootstrapped NLLS approach. Furthermore, we demonstrate how Bayesian posterior probability distributions can be employed to assess treatment response in a breast cancer DCE-MRI dataset using Cohen's d. Accuracy and precision of the BTM posterior distributions were validated and found to be in good agreement with the NLLS approaches, and assessment of therapy response with respect to uncertainty in parameter estimates was found to be excellent. In conclusion, the Bayesian modeling approach provides an elegant means to determine uncertainty via posterior distributions within a single step and provides honest information about changes in parameter estimates.
Collapse
Affiliation(s)
- Andreas Mittermeier
- Department of Radiology, Ludwig-Maximilians-University Hospital Munich, Munich, Germany. Author to whom any correspondence should be addressed
| | | | | | | | | |
Collapse
|
40
|
Wang S, Fan X, Zhang Y, Medved M, He D, Yousuf A, Jamison E, Oto A, Karczmar GS. Use of Indicator Dilution Principle to Evaluate Accuracy of Arterial Input Function Measured With Low-Dose Ultrafast Prostate Dynamic Contrast-Enhanced MRI. ACTA ACUST UNITED AC 2019; 5:260-265. [PMID: 31245547 PMCID: PMC6588202 DOI: 10.18383/j.tom.2019.00004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Accurately measuring arterial input function (AIF) is essential for quantitative analysis of dynamic contrast-enhanced (DCE) magnetic resonance imaging (MRI). We used the indicator dilution principle to evaluate the accuracy of AIF measured directly from an artery following a low-dose contrast media ultrafast DCE-MRI. In total, 15 patients with biopsy-confirmed localized prostate cancers were recruited. Cardiac MRI (CMRI) and ultrafast DCE-MRI were acquired on a Philips 3 T Ingenia scanner. The AIF was measured at iliac arties following injection of a low-dose (0.015 mmol/kg) gadolinium (Gd) contrast media. The cardiac output (CO) from CMRI (COCMRI) was calculated from the difference in ventricular volume at diastole and systole measured on the short axis of heart. The CO from DCE-MRI (CODCE) was also calculated from the AIF and dose of the contrast media used. A correlation test and Bland–Altman plot were used to compare COCMRI and CODCE. The average (±standard deviation [SD]) area under the curve measured directly from local AIF was 0.219 ± 0.07 mM·min. The average (±SD) COCMRI and CODCE were 6.52 ± 1.47 L/min and 6.88 ± 1.64 L/min, respectively. There was a strong positive correlation (r = 0.82, P < .01) and good agreement between COCMRI and CODCE. The CODCE is consistent with the reference standard COCMRI. This indicates that the AIF can be measured accurately from an artery with ultrafast DCE-MRI following injection of a low-dose contrast media.
Collapse
Affiliation(s)
- Shiyang Wang
- Department of Radiology, University of Chicago, Chicago, IL and
| | - Xiaobing Fan
- Department of Radiology, University of Chicago, Chicago, IL and
| | - Yue Zhang
- Department of Radiology, University of Chicago, Chicago, IL and
| | - Milica Medved
- Department of Radiology, University of Chicago, Chicago, IL and
| | - Dianning He
- Department of Radiology, University of Chicago, Chicago, IL and.,Sino-Dutch Biomedical and Information Engineering School, Northeastern University, Shenyang, China
| | - Ambereen Yousuf
- Department of Radiology, University of Chicago, Chicago, IL and
| | - Ernest Jamison
- Department of Radiology, University of Chicago, Chicago, IL and
| | - Aytekin Oto
- Department of Radiology, University of Chicago, Chicago, IL and
| | | |
Collapse
|
41
|
Rampun A, Scotney BW, Morrow PJ, Wang H, Winder J. Segmentation of breast MR images using a generalised 2D mathematical model with inflation and deflation forces of active contours. Artif Intell Med 2019; 97:44-60. [DOI: 10.1016/j.artmed.2018.10.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 09/26/2018] [Accepted: 10/23/2018] [Indexed: 11/28/2022]
|
42
|
Cao J, Pickup S, Clendenin C, Blouw B, Choi H, Kang D, Rosen M, O'Dwyer PJ, Zhou R. Dynamic Contrast-enhanced MRI Detects Responses to Stroma-directed Therapy in Mouse Models of Pancreatic Ductal Adenocarcinoma. Clin Cancer Res 2019; 25:2314-2322. [PMID: 30587546 PMCID: PMC6445712 DOI: 10.1158/1078-0432.ccr-18-2276] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 11/20/2018] [Accepted: 12/19/2018] [Indexed: 12/18/2022]
Abstract
PURPOSE The dense stroma underlies the drug resistance of pancreatic ductal adenocarcinoma (PDA) and has motivated the development of stroma-directed drugs. Our objective is to test the concept that dynamic contrast-enhanced (DCE) MRI using FDA-approved contrast media, an imaging method sensitive to the tumor microenvironment, can detect early responses to stroma-directed drug. EXPERIMENTAL DESIGN Imaging studies were performed in three mouse models exhibiting high desmoplastic reactions: the autochthonous PDA in genetically engineered mice (KPC), an orthotopic model in syngeneic mice, and a xenograft model of human PDA in athymic mice. An investigational drug, PEGPH20 (pegvorhyaluronidase alfa), which degrades hyaluronan (HA) in the stroma of PDA, was injected alone or in combination with gemcitabine. RESULTS At 24 hours after a single injection of PEGPH20, Ktrans , a DCE-MRI-derived marker that measures how fast a unit volume of contrast media is transferred from capillaries to interstitial space, increased 56% and 50% from baseline in the orthotopic and xenograft tumors, respectively, compared with a 4% and 6% decrease in vehicle groups (both P < 0.05). Similarly, after three combined treatments, Ktrans in KPC mice increased 54%, whereas it decreased 4% in controls treated with gemcitabine alone (P < 0.05). Consistently, after a single injection of PEGPH20, tumor HA content assessed by IHC was reduced substantially in all three models while drug delivery (measured by paclitaxel accumulation in tumor) was increased by 2.6-fold. CONCLUSIONS These data demonstrated a DCE-MRI marker, Ktrans , can detect early responses to stroma-directed drug and reveal the sustained effect of combination treatment (PEGPH20+ gemcitabine).
Collapse
Affiliation(s)
- Jianbo Cao
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania
- Medical College, Xiamen University, Xiamen, Fujian, P.R. China
| | - Stephen Pickup
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Cynthia Clendenin
- Pancreatic Cancer Research Center, University of Pennsylvania, Philadelphia, Pennsylvania
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Hoon Choi
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - David Kang
- Halozyme Therapeutics, San Diego, California
| | - Mark Rosen
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Peter J O'Dwyer
- Pancreatic Cancer Research Center, University of Pennsylvania, Philadelphia, Pennsylvania.
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Rong Zhou
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania.
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
43
|
Huang W, Chen Y, Fedorov A, Li X, Jajamovich GH, Malyarenko DI, Aryal MP, LaViolette PS, Oborski MJ, O'Sullivan F, Abramson RG, Jafari-Khouzani K, Afzal A, Tudorica A, Moloney B, Gupta SN, Besa C, Kalpathy-Cramer J, Mountz JM, Laymon CM, Muzi M, Kinahan PE, Schmainda K, Cao Y, Chenevert TL, Taouli B, Yankeelov TE, Fennessy F, Li X. The Impact of Arterial Input Function Determination Variations on Prostate Dynamic Contrast-Enhanced Magnetic Resonance Imaging Pharmacokinetic Modeling: A Multicenter Data Analysis Challenge, Part II. Tomography 2019; 5:99-109. [PMID: 30854447 PMCID: PMC6403046 DOI: 10.18383/j.tom.2018.00027] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
This multicenter study evaluated the effect of variations in arterial input function (AIF) determination on pharmacokinetic (PK) analysis of dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) data using the shutter-speed model (SSM). Data acquired from eleven prostate cancer patients were shared among nine centers. Each center used a site-specific method to measure the individual AIF from each data set and submitted the results to the managing center. These AIFs, their reference tissue-adjusted variants, and a literature population-averaged AIF, were used by the managing center to perform SSM PK analysis to estimate Ktrans (volume transfer rate constant), ve (extravascular, extracellular volume fraction), kep (efflux rate constant), and τi (mean intracellular water lifetime). All other variables, including the definition of the tumor region of interest and precontrast T1 values, were kept the same to evaluate parameter variations caused by variations in only the AIF. Considerable PK parameter variations were observed with within-subject coefficient of variation (wCV) values of 0.58, 0.27, 0.42, and 0.24 for Ktrans, ve, kep, and τi, respectively, using the unadjusted AIFs. Use of the reference tissue-adjusted AIFs reduced variations in Ktrans and ve (wCV = 0.50 and 0.10, respectively), but had smaller effects on kep and τi (wCV = 0.39 and 0.22, respectively). kep is less sensitive to AIF variation than Ktrans, suggesting it may be a more robust imaging biomarker of prostate microvasculature. With low sensitivity to AIF uncertainty, the SSM-unique τi parameter may have advantages over the conventional PK parameters in a longitudinal study.
Collapse
Affiliation(s)
- Wei Huang
- Oregon Health and Science University, Portland, OR
| | - Yiyi Chen
- Oregon Health and Science University, Portland, OR
| | - Andriy Fedorov
- Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Xia Li
- General Electric Global Research, Niskayuna, NY
| | | | | | | | | | | | | | | | | | - Aneela Afzal
- Oregon Health and Science University, Portland, OR
| | | | | | | | - Cecilia Besa
- Icahn School of Medicine at Mt Sinai, New York, NY
| | | | | | | | - Mark Muzi
- University of Washington, Seattle, WA; and
| | | | | | - Yue Cao
- University of Michigan, Ann Arbor, MI
| | | | | | | | - Fiona Fennessy
- Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Xin Li
- Oregon Health and Science University, Portland, OR
| |
Collapse
|
44
|
da Silva Neto OP, Araújo JDL, Caldas Oliveira AG, Cutrim M, Silva AC, Paiva AC, Gattass M. Pathophysiological mapping of tumor habitats in the breast in DCE-MRI using molecular texture descriptor. Comput Biol Med 2019; 106:114-125. [PMID: 30711799 DOI: 10.1016/j.compbiomed.2019.01.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 01/16/2019] [Accepted: 01/19/2019] [Indexed: 11/25/2022]
Abstract
BACKGROUND We propose a computational methodology capable of detecting and analyzing breast tumor habitats in images acquired by magnetic resonance imaging with dynamic contrast enhancement (DCE-MRI), based on the pathophysiological behavior of the contrast agent (CA). METHODS The proposed methodology comprises three steps. In summary, the first step is the acquisition of images from the Quantitative Imaging Network Breast. In the second step, the segmentation of the breasts is performed to remove the background, noise, and other unwanted objects from the image. In the third step, the generation of habitats is performed by applying two techniques: the molecular texture descriptor (MTD) that highlights the CA regions in the breast, and pathophysiological texture mapping (MPT), which generates tumor habitats based on the behavior of the CA. The combined use of these two techniques allows the automatic detection of tumors in the breast and analysis of each separate habitat with respect to their malignancy type. RESULTS The results found in this study were promising, with 100% of breast tumors being identified. The segmentation results exhibited an accuracy of 99.95%, sensitivity of 71.07%, specificity of 99.98%, and volumetric similarity of 77.75%. Moreover, we were able to classify the malignancy of the tumors, with 6 classified as malignant type III (WashOut) and 14 as malignant type II (Plateau), for a total of 20 cases. CONCLUSION We proposed a method for the automatic detection of tumors in the breast in DCE-MRI and performed the pathophysiological mapping of tumor habitats by analyzing the behavior of the CA, combining MTD and MPT, which allowed the mapping of internal tumor habitats.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Marcelo Gattass
- Pontifical Catholic University of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
45
|
Debus C, Floca R, Ingrisch M, Kompan I, Maier-Hein K, Abdollahi A, Nolden M. MITK-ModelFit: A generic open-source framework for model fits and their exploration in medical imaging - design, implementation and application on the example of DCE-MRI. BMC Bioinformatics 2019; 20:31. [PMID: 30651067 PMCID: PMC6335810 DOI: 10.1186/s12859-018-2588-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 12/19/2018] [Indexed: 01/21/2023] Open
Abstract
Background Many medical imaging techniques utilize fitting approaches for quantitative parameter estimation and analysis. Common examples are pharmacokinetic modeling in dynamic contrast-enhanced (DCE) magnetic resonance imaging (MRI)/computed tomography (CT), apparent diffusion coefficient calculations and intravoxel incoherent motion modeling in diffusion-weighted MRI and Z-spectra analysis in chemical exchange saturation transfer MRI. Most available software tools are limited to a special purpose and do not allow for own developments and extensions. Furthermore, they are mostly designed as stand-alone solutions using external frameworks and thus cannot be easily incorporated natively in the analysis workflow. Results We present a framework for medical image fitting tasks that is included in the Medical Imaging Interaction Toolkit MITK, following a rigorous open-source, well-integrated and operating system independent policy. Software engineering-wise, the local models, the fitting infrastructure and the results representation are abstracted and thus can be easily adapted to any model fitting task on image data, independent of image modality or model. Several ready-to-use libraries for model fitting and use-cases, including fit evaluation and visualization, were implemented. Their embedding into MITK allows for easy data loading, pre- and post-processing and thus a natural inclusion of model fitting into an overarching workflow. As an example, we present a comprehensive set of plug-ins for the analysis of DCE MRI data, which we validated on existing and novel digital phantoms, yielding competitive deviations between fit and ground truth. Conclusions Providing a very flexible environment, our software mainly addresses developers of medical imaging software that includes model fitting algorithms and tools. Additionally, the framework is of high interest to users in the domain of perfusion MRI, as it offers feature-rich, freely available, validated tools to perform pharmacokinetic analysis on DCE MRI data, with both interactive and automatized batch processing workflows.
Collapse
Affiliation(s)
- Charlotte Debus
- German Cancer Consortium (DKTK), Heidelberg, Germany. .,Department of Translational Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany. .,Department of Radiation Oncology, Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg University Hospital, Heidelberg, Germany. .,National Center for Tumor Diseases (NCT), Heidelberg, Germany. .,Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany.
| | - Ralf Floca
- Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany. .,Division of Medical Image Computing, German Cancer Research Center DKFZ, Heidelberg, Germany.
| | - Michael Ingrisch
- Department of Radiology, University Hospital Munich, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Ina Kompan
- Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany.,Division of Medical Image Computing, German Cancer Research Center DKFZ, Heidelberg, Germany
| | - Klaus Maier-Hein
- Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany.,Division of Medical Image Computing, German Cancer Research Center DKFZ, Heidelberg, Germany.,Section Pattern Recognition, Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Amir Abdollahi
- German Cancer Consortium (DKTK), Heidelberg, Germany.,Department of Translational Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Radiation Oncology, Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg University Hospital, Heidelberg, Germany.,National Center for Tumor Diseases (NCT), Heidelberg, Germany.,Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany
| | - Marco Nolden
- Division of Medical Image Computing, German Cancer Research Center DKFZ, Heidelberg, Germany
| |
Collapse
|
46
|
Hansen MB, Tietze A, Haack S, Kallehauge J, Mikkelsen IK, Østergaard L, Mouridsen K. Robust estimation of hemo-dynamic parameters in traditional DCE-MRI models. PLoS One 2019; 14:e0209891. [PMID: 30605459 PMCID: PMC6317807 DOI: 10.1371/journal.pone.0209891] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 11/09/2018] [Indexed: 01/04/2023] Open
Abstract
PURPOSE In dynamic contrast enhanced (DCE) MRI, separation of signal contributions from perfusion and leakage requires robust estimation of parameters in a pharmacokinetic model. We present and quantify the performance of a method to compute tissue hemodynamic parameters from DCE data using established pharmacokinetic models. METHODS We propose a Bayesian scheme to obtain perfusion metrics from DCE MRI data. Initial performance is assessed through digital phantoms of the extended Tofts model (ETM) and the two-compartment exchange model (2CXM), comparing the Bayesian scheme to the standard Levenberg-Marquardt (LM) algorithm. Digital phantoms are also invoked to identify limitations in the pharmacokinetic models related to measurement conditions. Using computed maps of the extra vascular volume (ve) from 19 glioma patients, we analyze differences in the number of un-physiological high-intensity ve values for both ETM and 2CXM, using a one-tailed paired t-test assuming un-equal variance. RESULTS The Bayesian parameter estimation scheme demonstrated superior performance over the LM technique in the digital phantom simulations. In addition, we identified limitations in parameter reliability in relation to scan duration for the 2CXM. DCE data for glioma and cervical cancer patients was analyzed with both algorithms and demonstrated improvement in image readability for the Bayesian method. The Bayesian method demonstrated significantly fewer non-physiological high-intensity ve values for the ETM (p<0.0001) and the 2CXM (p<0.0001). CONCLUSION We have demonstrated substantial improvement of the perceptive quality of pharmacokinetic parameters from advanced compartment models using the Bayesian parameter estimation scheme as compared to the LM technique.
Collapse
Affiliation(s)
- Mikkel B. Hansen
- Center of Functionally Integrative Neuroscience and MINDLab, Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Anna Tietze
- Center of Functionally Integrative Neuroscience and MINDLab, Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Inst. of Neuroradiology, Charité University Medicine Berlin, Berlin, Germany
| | - Søren Haack
- Department of Clinical Engineering, Aarhus University Hospital, Aarhus, Denmark
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Jesper Kallehauge
- Department of Medical Physics, Aarhus University Hospital, Aarhus, Denmark
| | - Irene K. Mikkelsen
- Center of Functionally Integrative Neuroscience and MINDLab, Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Leif Østergaard
- Center of Functionally Integrative Neuroscience and MINDLab, Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Medical Physics, Aarhus University Hospital, Aarhus, Denmark
| | - Kim Mouridsen
- Center of Functionally Integrative Neuroscience and MINDLab, Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
47
|
Jia K, Li L, Wu XJ, Hao MJ, Xue HY. Contrast-enhanced ultrasound for evaluating the pathologic response of breast cancer to neoadjuvant chemotherapy: A meta-analysis. Medicine (Baltimore) 2019; 98:e14258. [PMID: 30681622 PMCID: PMC6358361 DOI: 10.1097/md.0000000000014258] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
OBJECTIVE Recent reports have suggested that contrast-enhanced ultrasound (CEUS) can be used to monitor the pathologic responses of breast cancer (BC) to neoadjuvant chemotherapy (NAC); however, the diagnostic performance of CEUS in BC has yet to be confirmed. Thus, we conducted a meta-analysis of related studies to explore the relationship between CEUS and pathologic responses of BC to NAC. MATERIALS AND METHODS We searched PubMed, Embase, Web of Science, ScienceDirect, and China National Knowledge Infrastructure databases for studies published until September 31, 2018. Study-specific odds ratios (ORs) and 95% confidence intervals (CIs) were calculated, and then ORs with 95% CIs were pooled to estimate the prognostic role of CEUS for the pathologic responses of BC to NAC. RESULTS Pooled meta-analysis of the 9 eligible studies that included 424 patients indicated the high performance of CEUS for monitoring pathologic responses to NAC (OR = 31.83, 95% CI: 16.69-60.67, P < .001), with no significant heterogeneity (I = 0.0%, P = .529). The pooled sensitivity, specificity, positive likelihood ratio, negative likelihood ratio, and diagnostic odds ratio were 87% (95% CI: 0.81-0.92), 84% (95% CI: 0.74-0.91), 5.5 (95% CI: 3.3-9.2), 0.15 (95% CI: 0.10-0.23), and 36 (95% CI: 18-70), respectively. An area under the curve of 0.92 (95% CI: 0.89-0.94) suggests a high ability for prognostic detection. Although Begg's funnel plot (P = .057) indicated the presence of publication bias among the included studies, the trim-and-fill method verified the stability of the pooled outcomes. Sensitivity analysis suggested that the pooled OR was robust. CONCLUSION Our results suggest that CEUS has a high diagnostic performance for the pathologic responses of BC to NAC. Further and better-designed studies should be performed to verify the clinical applications of CEUS for monitoring BC responses to NAC.
Collapse
|
48
|
Parra NA, Lu H, Li Q, Stoyanova R, Pollack A, Punnen S, Choi J, Abdalah M, Lopez C, Gage K, Park JY, Kosj Y, Pow-Sang JM, Gillies RJ, Balagurunathan Y. Predicting clinically significant prostate cancer using DCE-MRI habitat descriptors. Oncotarget 2018; 9:37125-37136. [PMID: 30647849 PMCID: PMC6324677 DOI: 10.18632/oncotarget.26437] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Accepted: 11/16/2018] [Indexed: 12/16/2022] Open
Abstract
Prostate cancer diagnosis and treatment continues to be a major public health challenge. The heterogeneity of the disease is one of the major factors leading to imprecise diagnosis and suboptimal disease management. The improved resolution of functional multi-parametric magnetic resonance imaging (mpMRI) has shown promise to improve detection and characterization of the disease. Regions that subdivide the tumor based on Dynamic Contrast Enhancement (DCE) of mpMRI are referred to as DCE-Habitats in this study. The DCE defined perfusion curve patterns on the identified tumor habitat region are used to assess clinical significance. These perfusion curves were systematically quantified using seven features in association with the patient biopsy outcome and classifier models were built to find the best discriminating characteristics between clinically significant and insignificant prostate lesions defined by Gleason score (GS). Multivariable analysis was performed independently on one institution and validated on the other, using a multi-parametric feature model, based on DCE characteristics and ADC features. The models had an intra institution Area under the Receiver Operating Characteristic (AUC) of 0.82. Trained on Institution I and validated on the cohort from Institution II, the AUC was also 0.82 (sensitivity 0.68, specificity 0.95).
Collapse
Affiliation(s)
- N Andres Parra
- Department of Cancer Physiology, H.L. Moffitt Cancer Center, Tampa, FL, USA
| | - Hong Lu
- Department of Cancer Physiology, H.L. Moffitt Cancer Center, Tampa, FL, USA.,Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Qian Li
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Radka Stoyanova
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Alan Pollack
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Sanoj Punnen
- Department of Urology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jung Choi
- Department of Radiology, H.L. Moffitt Cancer Center, Tampa, FL, USA
| | - Mahmoud Abdalah
- Department of Cancer Physiology, H.L. Moffitt Cancer Center, Tampa, FL, USA
| | - Christopher Lopez
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Kenneth Gage
- Department of Radiology, H.L. Moffitt Cancer Center, Tampa, FL, USA
| | - Jong Y Park
- Department of Cancer Epidemiology, H.L. Moffitt Cancer Center, Tampa, FL, USA
| | - Yamoah Kosj
- Department of Cancer Epidemiology, H.L. Moffitt Cancer Center, Tampa, FL, USA.,Department of Radiation Oncology, H.L. Moffitt Cancer Center, Tampa, FL, USA
| | - Julio M Pow-Sang
- Department of Urology, H.L. Moffitt Cancer Center, Tampa, FL, USA
| | - Robert J Gillies
- Department of Cancer Physiology, H.L. Moffitt Cancer Center, Tampa, FL, USA.,Department of Radiology, H.L. Moffitt Cancer Center, Tampa, FL, USA
| | | |
Collapse
|
49
|
Shukla-Dave A, Obuchowski NA, Chenevert TL, Jambawalikar S, Schwartz LH, Malyarenko D, Huang W, Noworolski SM, Young RJ, Shiroishi MS, Kim H, Coolens C, Laue H, Chung C, Rosen M, Boss M, Jackson EF. Quantitative imaging biomarkers alliance (QIBA) recommendations for improved precision of DWI and DCE-MRI derived biomarkers in multicenter oncology trials. J Magn Reson Imaging 2018; 49:e101-e121. [PMID: 30451345 DOI: 10.1002/jmri.26518] [Citation(s) in RCA: 240] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 09/06/2018] [Accepted: 09/06/2018] [Indexed: 12/14/2022] Open
Abstract
Physiological properties of tumors can be measured both in vivo and noninvasively by diffusion-weighted imaging and dynamic contrast-enhanced magnetic resonance imaging. Although these techniques have been used for more than two decades to study tumor diffusion, perfusion, and/or permeability, the methods and studies on how to reduce measurement error and bias in the derived imaging metrics is still lacking in the literature. This is of paramount importance because the objective is to translate these quantitative imaging biomarkers (QIBs) into clinical trials, and ultimately in clinical practice. Standardization of the image acquisition using appropriate phantoms is the first step from a technical performance standpoint. The next step is to assess whether the imaging metrics have clinical value and meet the requirements for being a QIB as defined by the Radiological Society of North America's Quantitative Imaging Biomarkers Alliance (QIBA). The goal and mission of QIBA and the National Cancer Institute Quantitative Imaging Network (QIN) initiatives are to provide technical performance standards (QIBA profiles) and QIN tools for producing reliable QIBs for use in the clinical imaging community. Some of QIBA's development of quantitative diffusion-weighted imaging and dynamic contrast-enhanced QIB profiles has been hampered by the lack of literature for repeatability and reproducibility of the derived QIBs. The available research on this topic is scant and is not in sync with improvements or upgrades in MRI technology over the years. This review focuses on the need for QIBs in oncology applications and emphasizes the importance of the assessment of their reproducibility and repeatability. Level of Evidence: 5 Technical Efficacy Stage: 1 J. Magn. Reson. Imaging 2019;49:e101-e121.
Collapse
Affiliation(s)
- Amita Shukla-Dave
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Nancy A Obuchowski
- Department of Quantitative Health Sciences, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Thomas L Chenevert
- Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Sachin Jambawalikar
- Department of Radiology, Columbia University Irving Medical Center, New York, New York, USA
| | - Lawrence H Schwartz
- Department of Radiology, Columbia University Irving Medical Center, New York, New York, USA
| | - Dariya Malyarenko
- Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Wei Huang
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, Oregon, USA
| | - Susan M Noworolski
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California, USA
| | - Robert J Young
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Mark S Shiroishi
- Division of Neuroradiology, Department of Radiology, University of Southern California, Los Angeles, California, USA
| | - Harrison Kim
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Catherine Coolens
- Department of Radiation Oncology, Princess Margaret Cancer Centre, Toronto, Canada
| | | | - Caroline Chung
- Department of Radiation Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Mark Rosen
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Michael Boss
- Applied Physics Division, National Institute of Standards and Technology, Boulder, Colorado, USA
| | - Edward F Jackson
- Departments of Medical Physics, Radiology, and Human Oncology, University of Wisconsin School of Medicine, Madison, Wisconsin, USA
| |
Collapse
|
50
|
Artzi M, Liberman G, Blumenthal DT, Bokstein F, Aizenstein O, Ben Bashat D. Repeatability of dynamic contrast enhanced v p parameter in healthy subjects and patients with brain tumors. J Neurooncol 2018; 140:727-737. [PMID: 30392091 DOI: 10.1007/s11060-018-03006-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 09/20/2018] [Indexed: 12/12/2022]
Abstract
PURPOSE To study the repeatability of plasma volume (vp) extracted from dynamic-contrast-enhanced (DCE) MRI in order to define threshold values for significant longitudinal changes, and to assess changes in patients with high-grade-glioma (HGG). METHODS Twenty eight healthy subjects, of which eleven scanned twice, were used to assess the repeatability of vp within the normal-appearing brain tissue and to define threshold values for significant changes based on least-detected-differences (LDD) of mean vp values and histogram comparisons using earth-mover's-distance (EMD). Sixteen patients with HGG were scanned longitudinally with eight patients scanned before and following bevacizumab therapy. Longitudinal changes were assessed based on defined threshold values in comparison to RANO criteria. RESULTS The threshold values for significant changes were: LDD = 0.0024 (ml/100 ml, 21%) for mean vp and EMD = 4.14. In patients, in 20/24 comparisons, no significant longitudinal changes were detected for vp within the normal-appearing brain tissue. Concurring results were obtained between changes in lesion volume (RANO criteria) and LDD or EMD values in cases diagnosed with progressive-disease, yet in about 50% of cases diagnosed with partial-response preliminary results demonstrated significant increase in vp despite significant reductions in lesion volume. In two patients, these changes preceded progression detected at follow-up scans. In general, a good concordance was obtained between LDD and EMD. CONCLUSION This study shows high repeatability of vp and provides threshold values for significant changes in longitudinal assessment of patients with brain tumors. Preliminary results suggest the use of vp-DCE parameter to improve assessment of therapy response in patients with high-grade-glioma.
Collapse
Affiliation(s)
- Moran Artzi
- Sagol Brain Institute, Tel Aviv Sourasky Medical Center, Tel-Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Gilad Liberman
- Sagol Brain Institute, Tel Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | - Deborah T Blumenthal
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Neuro-Oncology Service, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Felix Bokstein
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Neuro-Oncology Service, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Orna Aizenstein
- Sagol Brain Institute, Tel Aviv Sourasky Medical Center, Tel-Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Dafna Ben Bashat
- Sagol Brain Institute, Tel Aviv Sourasky Medical Center, Tel-Aviv, Israel. .,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel. .,Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|