1
|
Bakas S, Vollmuth P, Galldiks N, Booth TC, Aerts HJWL, Bi WL, Wiestler B, Tiwari P, Pati S, Baid U, Calabrese E, Lohmann P, Nowosielski M, Jain R, Colen R, Ismail M, Rasool G, Lupo JM, Akbari H, Tonn JC, Macdonald D, Vogelbaum M, Chang SM, Davatzikos C, Villanueva-Meyer JE, Huang RY. Artificial Intelligence for Response Assessment in Neuro Oncology (AI-RANO), part 2: recommendations for standardisation, validation, and good clinical practice. Lancet Oncol 2024; 25:e589-e601. [PMID: 39481415 DOI: 10.1016/s1470-2045(24)00315-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 05/21/2024] [Accepted: 05/23/2024] [Indexed: 11/02/2024]
Abstract
Technological advancements have enabled the extended investigation, development, and application of computational approaches in various domains, including health care. A burgeoning number of diagnostic, predictive, prognostic, and monitoring biomarkers are continuously being explored to improve clinical decision making in neuro-oncology. These advancements describe the increasing incorporation of artificial intelligence (AI) algorithms, including the use of radiomics. However, the broad applicability and clinical translation of AI are restricted by concerns about generalisability, reproducibility, scalability, and validation. This Policy Review intends to serve as the leading resource of recommendations for the standardisation and good clinical practice of AI approaches in health care, particularly in neuro-oncology. To this end, we investigate the repeatability, reproducibility, and stability of AI in response assessment in neuro-oncology in studies on factors affecting such computational approaches, and in publicly available open-source data and computational software tools facilitating these goals. The pathway for standardisation and validation of these approaches is discussed with the view of trustworthy AI enabling the next generation of clinical trials. We conclude with an outlook on the future of AI-enabled neuro-oncology.
Collapse
Affiliation(s)
- Spyridon Bakas
- Department of Pathology & Laboratory Medicine, Division of Computational Pathology, Indiana University, Indianopolis, IN, USA; Department of Radiology & Imaging Sciences, School of Medicine, Indiana University, Indianapolis, IN, USA; Department of Neurological Surgery, School of Medicine, Indiana University, Indianapolis, IN, USA; Department of Biostatistics and Health Data Science, School of Medicine, Indiana University, Indianapolis, IN, USA; Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianopolis, IN, USA; Department of Computer Science, Luddy School of Informatics, Computing, and Engineering, Indiana University, Indianapolis, IN, USA.
| | - Philipp Vollmuth
- Division for Computational Radiology and Clinical AI, Clinic for Neuroradiology, University Hospital Bonn, Bonn, Germany; Faculty of Medicine, University of Bonn, Bonn, Germany; Division for Medical Image Computing, German Cancer Research Center, Heidelberg, Germany
| | - Norbert Galldiks
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, Cologne, Germany; Institute of Neuroscience and Medicine, Research Center Juelich, Juelich, Germany
| | - Thomas C Booth
- School of Biomedical Engineering & Imaging Sciences, King's College London, London, UK; Department of Neuroradiology, King's College Hospital NHS Foundation Trust, London, UK
| | - Hugo J W L Aerts
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Artificial Intelligence in Medicine Program, Mass General Brigham, Harvard Medical School, Boston, MA, USA; Radiology and Nuclear Medicine, Maastricht University, Maastricht, Netherlands
| | - Wenya Linda Bi
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Benedikt Wiestler
- Department of Neuroradiology, University Hospital, Technical University of Munich, Munich, Germany
| | - Pallavi Tiwari
- Department of Radiology, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Sarthak Pati
- Department of Pathology & Laboratory Medicine, Division of Computational Pathology, Indiana University, Indianopolis, IN, USA
| | - Ujjwal Baid
- Department of Pathology & Laboratory Medicine, Division of Computational Pathology, Indiana University, Indianopolis, IN, USA; Department of Radiology & Imaging Sciences, School of Medicine, Indiana University, Indianapolis, IN, USA; Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianopolis, IN, USA
| | - Evan Calabrese
- Department of Radiology, School of Medicine, Duke University, Durham, NC, USA
| | - Philipp Lohmann
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, Cologne, Germany; Department of Nuclear Medicine, University Hospital RWTH Aachen, Aachen, Germany
| | - Martha Nowosielski
- Department of Neurology, Medical University Innsbruck, Innsbruck, Austria
| | - Rajan Jain
- Department of Radiology and Department of Neurosurgery, NYU Grossman School of Medicine, New York, NY, USA
| | - Rivka Colen
- Department of Radiology, Neuroradiology Division, Center for Artificial Intelligence Innovation in Medical Imaging, University of Pittsburgh, Pittsburgh, PA, USA
| | - Marwa Ismail
- Department of Radiology, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Ghulam Rasool
- Department of Machine Learning, H Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Janine M Lupo
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Hamed Akbari
- Department of Bioengineering, School of Engineering, Santa Clara University, Santa Clara, CA, USA
| | - Joerg C Tonn
- Department of Neurosurgery, Ludwig-Maximilians-University, Munich, Germany; German Cancer Consortium, Partner Site Munich, Munich, Germany
| | | | - Michael Vogelbaum
- Department of Neuro-Oncology, H Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA; Department of Neurosurgery, H Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA; H Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Susan M Chang
- Department of Neurological Surgery, Division of Neuro-Oncology, University of California San Francisco, San Francisco, CA, USA
| | - Christos Davatzikos
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Center for Artificial Intelligence for Integrated Diagnostics and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Javier E Villanueva-Meyer
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Raymond Y Huang
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
2
|
Villanueva-Meyer JE, Bakas S, Tiwari P, Lupo JM, Calabrese E, Davatzikos C, Bi WL, Ismail M, Akbari H, Lohmann P, Booth TC, Wiestler B, Aerts HJWL, Rasool G, Tonn JC, Nowosielski M, Jain R, Colen RR, Pati S, Baid U, Vollmuth P, Macdonald D, Vogelbaum MA, Chang SM, Huang RY, Galldiks N. Artificial Intelligence for Response Assessment in Neuro Oncology (AI-RANO), part 1: review of current advancements. Lancet Oncol 2024; 25:e581-e588. [PMID: 39481414 DOI: 10.1016/s1470-2045(24)00316-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 05/21/2024] [Accepted: 05/23/2024] [Indexed: 11/02/2024]
Abstract
The development, application, and benchmarking of artificial intelligence (AI) tools to improve diagnosis, prognostication, and therapy in neuro-oncology are increasing at a rapid pace. This Policy Review provides an overview and critical assessment of the work to date in this field, focusing on diagnostic AI models of key genomic markers, predictive AI models of response before and after therapy, and differentiation of true disease progression from treatment-related changes, which is a considerable challenge based on current clinical care in neuro-oncology. Furthermore, promising future directions, including the use of AI for automated response assessment in neuro-oncology, are discussed.
Collapse
Affiliation(s)
- Javier E Villanueva-Meyer
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA; Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA.
| | - Spyridon Bakas
- Division of Computational Pathology, Department of Pathology & Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Radiology & Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Biostatistics & Health Data Science, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA; Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN, USA; Department of Computer Science, Luddy School of Informatics, Computing, and Engineering, Indiana University, Indianapolis, IN, USA
| | - Pallavi Tiwari
- Department of Radiology and Biomedical Engineering, University of Wisconsin, Madison, WI, USA
| | - Janine M Lupo
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Evan Calabrese
- Duke University Center for Artificial Intelligence in Radiology, Department of Radiology, Duke University, Durham, NC, USA
| | - Christos Davatzikos
- Center for Artificial Intelligence and Data Science for Integrated Diagnostics (AI2D) and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA; Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Wenya Linda Bi
- Department of Neurosurgery, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Marwa Ismail
- Department of Radiology and Biomedical Engineering, University of Wisconsin, Madison, WI, USA
| | - Hamed Akbari
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Bioengineering, Santa Clara University, Santa Clara, CA, USA
| | - Philipp Lohmann
- Institute of Neuroscience and Medicine (INM-4), Research Center Juelich (FZJ), Juelich, Germany; Department of Nuclear Medicine, University Hospital RWTH Aachen, Aachen, Germany
| | - Thomas C Booth
- Department of Neuroradiology, Ruskin Wing, King's College Hospital NHS Foundation Trust, London, UK; School of Biomedical Engineering & Imaging Sciences, King's College London, London, UK; London Regional Cancer Program, London, UK
| | - Benedikt Wiestler
- Department of Neuroradiology, University Hospital, Technical University of Munich, Munich, Germany
| | - Hugo J W L Aerts
- Artificial Intelligence in Medicine (AIM) Program, Mass General Brigham, Harvard Medical School, Boston, MA, USA; Radiology and Nuclear Medicine, CARIM & GROW, Maastricht University, Maastricht, Netherlands
| | - Ghulam Rasool
- Department of Machine Learning, H Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Joerg C Tonn
- Department of Neurosurgery, Ludwig Maximilians University, Munich, Germany and German Cancer Consortium (DKTK), Partner Site Munich, Germany
| | - Martha Nowosielski
- Department of Neurology, Medical University Innsbruck, Innsbruck, Austria
| | - Rajan Jain
- Department of Radiology and Department of Neurosurgery, New York University Langone Health, New York, NY, USA
| | - Rivka R Colen
- Radiology and Nuclear Medicine, CARIM & GROW, Maastricht University, Maastricht, Netherlands
| | - Sarthak Pati
- Division of Computational Pathology, Department of Pathology & Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ujjwal Baid
- Division of Computational Pathology, Department of Pathology & Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Philipp Vollmuth
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | - David Macdonald
- Department of Neuro-Oncology, H Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Michael A Vogelbaum
- Department of Neurosurgery, H Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA; Department of Machine Learning, H Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Susan M Chang
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Raymond Y Huang
- Department of Radiology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Norbert Galldiks
- Institute of Neuroscience and Medicine (INM-4), Research Center Juelich (FZJ), Juelich, Germany; Department of Neurology, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| |
Collapse
|
3
|
Yan RE, Greenfield JP. Challenges and Outlooks in Precision Medicine: Expectations Versus Reality. World Neurosurg 2024; 190:573-581. [PMID: 39425299 DOI: 10.1016/j.wneu.2024.06.142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 10/21/2024]
Abstract
Recent developments in technology have led to rapid advances in precision medicine, especially due to the rise of next-generation sequencing and molecular profiling. These technological advances have led to rapid advances in research, including increased tumor subtype resolution, new therapeutic agents, and mechanistic insights. Certain therapies have even been approved for molecular biomarkers across histopathological diagnoses; however, translation of research findings to the clinic still faces a number of challenges. In this review, the authors discuss several key challenges to the clinical integration of precision medicine, including the blood-brain barrier, both a lack and excess of molecular targets, and tumor heterogeneity/escape from therapy. They also highlight a few key efforts to address these challenges, including new frontiers in drug delivery, a rapidly expanding treatment repertoire, and improvements in active response monitoring. With continued improvements and developments, the authors anticipate that precision medicine will increasingly become the gold standard for clinical care.
Collapse
Affiliation(s)
- Rachel E Yan
- Department of Neurological Surgery, Weill Cornell Medicine, New York, New York, USA
| | - Jeffrey P Greenfield
- Department of Neurological Surgery, NewYork-Presbyterian Weill Cornell Medicine, New York, New York, USA.
| |
Collapse
|
4
|
Liu Y, Weng L, Wang Y, Zhang J, Wu Q, Zhao P, Shi Y, Wang P, Fang L. Deciphering the role of CD47 in cancer immunotherapy. J Adv Res 2024; 63:129-158. [PMID: 39167629 PMCID: PMC11380025 DOI: 10.1016/j.jare.2023.10.009] [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: 06/24/2023] [Revised: 10/05/2023] [Accepted: 10/18/2023] [Indexed: 08/23/2024] Open
Abstract
BACKGROUND Immunotherapy has emerged as a novel strategy for cancer treatment following surgery, radiotherapy, and chemotherapy. Immune checkpoint blockade and Chimeric antigen receptor (CAR)-T cell therapies have been successful in clinical trials. Cancer cells evade immune surveillance by hijacking inhibitory pathways via overexpression of checkpoint genes. The Cluster of Differentiation 47 (CD47) has emerged as a crucial checkpoint for cancer immunotherapy by working as a "don't eat me" signal and suppressing innate immune signaling. Furthermore, CD47 is highly expressed in many cancer types to protect cancer cells from phagocytosis via binding to SIRPα on phagocytes. Targeting CD47 by either interrupting the CD47-SIRPα axis or combing with other therapies has been demonstrated as an encouraging therapeutic strategy in cancer immunotherapy. Antibodies and small molecules that target CD47 have been explored in pre- and clinical trials. However, formidable challenges such as the anemia and palate aggregation cannot be avoided because of the wide presentation of CD47 on erythrocytes. AIM OF VIEW This review summarizes the current knowledge on the regulation and function of CD47, and provides a new perspective for immunotherapy targeting CD47. It also highlights the clinical progress of targeting CD47 and discusses challenges and potential strategies. KEY SCIENTIFIC CONCEPTS OF REVIEW This review provides a comprehensive understanding of targeting CD47 in cancer immunotherapy, it also augments the concept of combination immunotherapy strategies by employing both innate and adaptive immune responses.
Collapse
Affiliation(s)
- Yu'e Liu
- Tongji University Cancer Center, Shanghai Tenth People's Hospital of Tongji University, School of Medicine, Tongji University, Shanghai 200092, China
| | - Linjun Weng
- Tongji University Cancer Center, Shanghai Tenth People's Hospital of Tongji University, School of Medicine, Tongji University, Shanghai 200092, China
| | - Yanjin Wang
- Department of Nephrology, Shanghai East Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Jin Zhang
- Department of Pharmacology and Toxicology, University of Mississippi, Medical Center, 39216 Jackson, MS, USA
| | - Qi Wu
- Tongji University Cancer Center, Shanghai Tenth People's Hospital of Tongji University, School of Medicine, Tongji University, Shanghai 200092, China
| | - Pengcheng Zhao
- School of Life Sciences and Medicine, Shandong University of Technology, No.266 Xincun West Road, Zibo 255000, Shandong Province, China
| | - Yufeng Shi
- Tongji University Cancer Center, Shanghai Tenth People's Hospital of Tongji University, School of Medicine, Tongji University, Shanghai 200092, China; Clinical Center for Brain and Spinal Cord Research, Tongji University, Shanghai 200092, China.
| | - Ping Wang
- Tongji University Cancer Center, Shanghai Tenth People's Hospital of Tongji University, School of Medicine, Tongji University, Shanghai 200092, China.
| | - Lan Fang
- Tongji University Cancer Center, Shanghai Tenth People's Hospital of Tongji University, School of Medicine, Tongji University, Shanghai 200092, China.
| |
Collapse
|
5
|
Yang Y, Luo X, Wang Y, Xu A, Peng L, Zhang X, Wang Z, Ying Y, Li K. β-Mangostin targets and suppresses glioma via STING activation and tumor-associated microglia polarization. Biomed Pharmacother 2024; 177:117074. [PMID: 38972149 DOI: 10.1016/j.biopha.2024.117074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 06/20/2024] [Accepted: 06/29/2024] [Indexed: 07/09/2024] Open
Abstract
Glioma, a common and highly malignant central nervous system tumor, markedly influences patient prognosis via interactions with glioma-associated macrophages. Previous research has revealed the anticancer potential of β-mangostin, a xanthone derivative obtained from the mangosteen fruit. This research investigated the role of β-mangostin on microglia in the glioma microenvironment and evaluated the efficacy of β-mangostin combined with anti-PD-1 antibody (αPD-1) in glioma-bearing mice. The results showed that, β-mangostin attenuated M2 polarization in BV2 cells and promoted M1-related interleukin (IL)-1β and IL-6 secretion, thereby inhibiting glioma invasion. In addition, β-mangostin improved the anti-glioma effects of αPD-1 and increased CD8+T cell and M1-type microglia infiltration. Mechanistically, β-mangostin bound to the stimulator of interferon genes (STING) protein, which is crucial for the anti-tumor innate immune response, and promoted STING phosphorylation in microglia, both in vivo and in vitro. These results provide insights into its mode of action and supporting further investigation into β-mangostin as a therapeutic agent.
Collapse
Affiliation(s)
- Yimin Yang
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Xuling Luo
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Yaling Wang
- Center for Laboratory Medicine, Allergy center, Department of Transfusion medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Aibo Xu
- Center for Laboratory Medicine, Allergy center, Department of Transfusion medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Lina Peng
- Center for Laboratory Medicine, Allergy center, Department of Transfusion medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Xiaoting Zhang
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Zhen Wang
- Center for Laboratory Medicine, Allergy center, Department of Transfusion medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang 310014, China; Key Laboratory of Biomarkers and In Vitro Diagnosis Translation of Zhejiang Province, Hangzhou, Zhejiang 310063, China.
| | - Youmin Ying
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China.
| | - Kaiqiang Li
- Center for Laboratory Medicine, Allergy center, Department of Transfusion medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang 310014, China; Key Laboratory of Biomarkers and In Vitro Diagnosis Translation of Zhejiang Province, Hangzhou, Zhejiang 310063, China.
| |
Collapse
|
6
|
Kersch CN, Kim M, Stoller J, Barajas RF, Park JE. Imaging Genomics of Glioma Revisited: Analytic Methods to Understand Spatial and Temporal Heterogeneity. AJNR Am J Neuroradiol 2024; 45:537-548. [PMID: 38548303 PMCID: PMC11288537 DOI: 10.3174/ajnr.a8148] [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: 07/28/2023] [Accepted: 11/09/2023] [Indexed: 04/12/2024]
Abstract
An improved understanding of the cellular and molecular biologic processes responsible for brain tumor development, growth, and resistance to therapy is fundamental to improving clinical outcomes. Imaging genomics is the study of the relationships between microscopic, genetic, and molecular biologic features and macroscopic imaging features. Imaging genomics is beginning to shift clinical paradigms for diagnosing and treating brain tumors. This article provides an overview of imaging genomics in gliomas, in which imaging data including hallmarks such as IDH-mutation, MGMT methylation, and EGFR-mutation status can provide critical insights into the pretreatment and posttreatment stages. This article will accomplish the following: 1) review the methods used in imaging genomics, including visual analysis, quantitative analysis, and radiomics analysis; 2) recommend suitable analytic methods for imaging genomics according to biologic characteristics; 3) discuss the clinical applicability of imaging genomics; and 4) introduce subregional tumor habitat analysis with the goal of guiding future radiogenetics research endeavors toward translation into critically needed clinical applications.
Collapse
Affiliation(s)
- Cymon N Kersch
- From the Department of Radiation Medicine (C.N.K.), Oregon Health and Science University, Portland, Oregon
| | - Minjae Kim
- Department of Radiology and Research Institute of Radiology (M.K., J.E.P.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jared Stoller
- Department of Diagnostic Radiology (J.S., R.F.B.), Oregon Health and Science University, Portland, Oregon
| | - Ramon F Barajas
- Department of Diagnostic Radiology (J.S., R.F.B.), Oregon Health and Science University, Portland, Oregon
- Knight Cancer Institute (R.F.B.), Oregon Health and Science University, Portland, Oregon
- Advanced Imaging Research Center (R.F.B.), Oregon Health and Science University, Portland, Oregon
| | - Ji Eun Park
- Department of Radiology and Research Institute of Radiology (M.K., J.E.P.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
7
|
Usuzaki T, Takahashi K, Inamori R, Morishita Y, Shizukuishi T, Takagi H, Ishikuro M, Obara T, Takase K. Identifying key factors for predicting O6-Methylguanine-DNA methyltransferase status in adult patients with diffuse glioma: a multimodal analysis of demographics, radiomics, and MRI by variable Vision Transformer. Neuroradiology 2024; 66:761-773. [PMID: 38472373 PMCID: PMC11031474 DOI: 10.1007/s00234-024-03329-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 03/04/2024] [Indexed: 03/14/2024]
Abstract
PURPOSE This study aimed to perform multimodal analysis by vision transformer (vViT) in predicting O6-methylguanine-DNA methyl transferase (MGMT) promoter status among adult patients with diffuse glioma using demographics (sex and age), radiomic features, and MRI. METHODS The training and test datasets contained 122 patients with 1,570 images and 30 patients with 484 images, respectively. The radiomic features were extracted from enhancing tumors (ET), necrotic tumor cores (NCR), and the peritumoral edematous/infiltrated tissues (ED) using contrast-enhanced T1-weighted images (CE-T1WI) and T2-weighted images (T2WI). The vViT had 9 sectors; 1 demographic sector, 6 radiomic sectors (CE-T1WI ET, CE-T1WI NCR, CE-T1WI ED, T2WI ET, T2WI NCR, and T2WI ED), 2 image sectors (CE-T1WI, and T2WI). Accuracy and area under the curve of receiver-operating characteristics (AUC-ROC) were calculated for the test dataset. The performance of vViT was compared with AlexNet, GoogleNet, VGG16, and ResNet by McNemar and Delong test. Permutation importance (PI) analysis with the Mann-Whitney U test was performed. RESULTS The accuracy was 0.833 (95% confidence interval [95%CI]: 0.714-0.877) and the area under the curve of receiver-operating characteristics was 0.840 (0.650-0.995) in the patient-based analysis. The vViT had higher accuracy than VGG16 and ResNet, and had higher AUC-ROC than GoogleNet (p<0.05). The ED radiomic features extracted from the T2-weighted image demonstrated the highest importance (PI=0.239, 95%CI: 0.237-0.240) among all other sectors (p<0.0001). CONCLUSION The vViT is a competent deep learning model in predicting MGMT status. The ED radiomic features of the T2-weighted image demonstrated the most dominant contribution.
Collapse
Affiliation(s)
- Takuma Usuzaki
- Department of Diagnostic Radiology, Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, Miyagi, 980-8574, Japan.
| | - Kengo Takahashi
- Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, Miyagi, 980-8573, Japan
| | - Ryusei Inamori
- Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, Miyagi, 980-8573, Japan
| | - Yohei Morishita
- Department of Diagnostic Radiology, Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, Miyagi, 980-8574, Japan
| | - Takashi Shizukuishi
- Department of Diagnostic Radiology, Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, Miyagi, 980-8574, Japan
| | - Hidenobu Takagi
- Department of Diagnostic Radiology, Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, Miyagi, 980-8574, Japan
- Department of Advanced MRI Collaborative Research, Graduate School of Medicine, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, Miyagi, 980-8573, Japan
| | - Mami Ishikuro
- Tohoku University Graduate School of Medicine, Division of Molecular Epidemiology, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, Miyagi, 980-8573, Japan
| | - Taku Obara
- Tohoku University Graduate School of Medicine, Division of Molecular Epidemiology, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, Miyagi, 980-8573, Japan
- Tohoku University Graduate School of Medicine, Division of Molecular Epidemiology, Department of Preventive Medicine and Epidemiology, Tohoku Medical Megabank Organization, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, Miyagi, 980-8573, Japan
- Tohoku University Hospital, Department of Pharmaceutical Sciences, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, Miyagi, 980-8574, Japan
| | - Kei Takase
- Department of Diagnostic Radiology, Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, Miyagi, 980-8574, Japan
| |
Collapse
|
8
|
Bhattacharya K, Rastogi S, Mahajan A. Post-treatment imaging of gliomas: challenging the existing dogmas. Clin Radiol 2024; 79:e376-e392. [PMID: 38123395 DOI: 10.1016/j.crad.2023.11.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 10/23/2023] [Accepted: 11/21/2023] [Indexed: 12/23/2023]
Abstract
Gliomas are the commonest malignant central nervous system tumours in adults and imaging is the cornerstone of diagnosis, treatment, and post-treatment follow-up of these patients. With the ever-evolving treatment strategies post-treatment imaging and interpretation in glioma remains challenging, more so with the advent of anti-angiogenic drugs and immunotherapy, which can significantly alter the appearance in this setting, thus making interpretation of routine imaging findings such as contrast enhancement, oedema, and mass effect difficult to interpret. This review details the various methods of management of glioma including the upcoming novel therapies and their impact on imaging findings, with a comprehensive description of the imaging findings in conventional and advanced imaging techniques. A systematic appraisal for the existing and emerging techniques of imaging in these settings and their clinical application including various response assessment guidelines and artificial intelligence based response assessment will also be discussed.
Collapse
Affiliation(s)
- K Bhattacharya
- Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - S Rastogi
- Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - A Mahajan
- Department of imaging, The Clatterbridge Cancer Centre, NHS Foundation Trust, Pembroke Place, Liverpool L7 8YA, UK; University of Liverpool, Liverpool L69 3BX, UK.
| |
Collapse
|
9
|
Rabah N, Ait Mohand FE, Kravchenko-Balasha N. Understanding Glioblastoma Signaling, Heterogeneity, Invasiveness, and Drug Delivery Barriers. Int J Mol Sci 2023; 24:14256. [PMID: 37762559 PMCID: PMC10532387 DOI: 10.3390/ijms241814256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 09/13/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
The most prevalent and aggressive type of brain cancer, namely, glioblastoma (GBM), is characterized by intra- and inter-tumor heterogeneity and strong spreading capacity, which makes treatment ineffective. A true therapeutic answer is still in its infancy despite various studies that have made significant progress toward understanding the mechanisms behind GBM recurrence and its resistance. The primary causes of GBM recurrence are attributed to the heterogeneity and diffusive nature; therefore, monitoring the tumor's heterogeneity and spreading may offer a set of therapeutic targets that could improve the clinical management of GBM and prevent tumor relapse. Additionally, the blood-brain barrier (BBB)-related poor drug delivery that prevents effective drug concentrations within the tumor is discussed. With a primary emphasis on signaling heterogeneity, tumor infiltration, and computational modeling of GBM, this review covers typical therapeutic difficulties and factors contributing to drug resistance development and discusses potential therapeutic approaches.
Collapse
Affiliation(s)
| | | | - Nataly Kravchenko-Balasha
- The Institute of Biomedical and Oral Research, Hebrew University of Jerusalem, Jerusalem 91120, Israel; (N.R.); (F.-E.A.M.)
| |
Collapse
|
10
|
Baheti B, Pati S, Menze B, Bakas S. Leveraging 2D Deep Learning ImageNet-trained models for Native 3D Medical Image Analysis. BRAINLESION : GLIOMA, MULTIPLE SCLEROSIS, STROKE AND TRAUMATIC BRAIN INJURIES. BRAINLES (WORKSHOP) 2023; 13769:68-79. [PMID: 37928819 PMCID: PMC10623403 DOI: 10.1007/978-3-031-33842-7_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2023]
Abstract
Convolutional neural networks (CNNs) have shown promising performance in various 2D computer vision tasks due to availability of large amounts of 2D training data. Contrarily, medical imaging deals with 3D data and usually lacks the equivalent extent and diversity of data, for developing AI models. Transfer learning provides the means to use models trained for one application as a starting point to another application. In this work, we leverage 2D pre-trained models as a starting point in 3D medical applications by exploring the concept of Axial-Coronal-Sagittal (ACS) convolutions. We have incorporated ACS as an alternative of native 3D convolutions in the Generally Nuanced Deep Learning Framework (GaNDLF), providing various well-established and state-of-the-art network architectures with the availability of pre-trained encoders from 2D data. Results of our experimental evaluation on 3D MRI data of brain tumor patients for i) tumor segmentation and ii) radiogenomic classification, show model size reduction by ~22% and improvement in validation accuracy by ~33%. Our findings support the advantage of ACS convolutions in pre-trained 2D CNNs over 3D CNN without pre-training, for 3D segmentation and classification tasks, democratizing existing models trained in datasets of unprecedented size and showing promise in the field of healthcare.
Collapse
Affiliation(s)
- Bhakti Baheti
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sarthak Pati
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Informatics, Technical University of Munich, Munich, Germany
| | - Bjoern Menze
- Department of Informatics, Technical University of Munich, Munich, Germany
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
| | - Spyridon Bakas
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
11
|
Rezaeijo SM, Chegeni N, Baghaei Naeini F, Makris D, Bakas S. Within-Modality Synthesis and Novel Radiomic Evaluation of Brain MRI Scans. Cancers (Basel) 2023; 15:3565. [PMID: 37509228 PMCID: PMC10377568 DOI: 10.3390/cancers15143565] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 06/27/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023] Open
Abstract
One of the most common challenges in brain MRI scans is to perform different MRI sequences depending on the type and properties of tissues. In this paper, we propose a generative method to translate T2-Weighted (T2W) Magnetic Resonance Imaging (MRI) volume from T2-weight-Fluid-attenuated-Inversion-Recovery (FLAIR) and vice versa using Generative Adversarial Networks (GAN). To evaluate the proposed method, we propose a novel evaluation schema for generative and synthetic approaches based on radiomic features. For the evaluation purpose, we consider 510 pair-slices from 102 patients to train two different GAN-based architectures Cycle GAN and Dual Cycle-Consistent Adversarial network (DC2Anet). The results indicate that generative methods can produce similar results to the original sequence without significant change in the radiometric feature. Therefore, such a method can assist clinics to make decisions based on the generated image when different sequences are not available or there is not enough time to re-perform the MRI scans.
Collapse
Affiliation(s)
- Seyed Masoud Rezaeijo
- Department of Medical Physics, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; (S.M.R.)
| | - Nahid Chegeni
- Department of Medical Physics, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; (S.M.R.)
| | - Fariborz Baghaei Naeini
- Faculty of Engineering, Computing and the Environment, Kingston University, Penrhyn Road Campus, Kingston upon Thames, London KT1 2EE, UK; (F.B.N.); (D.M.)
| | - Dimitrios Makris
- Faculty of Engineering, Computing and the Environment, Kingston University, Penrhyn Road Campus, Kingston upon Thames, London KT1 2EE, UK; (F.B.N.); (D.M.)
| | - Spyridon Bakas
- Faculty of Engineering, Computing and the Environment, Kingston University, Penrhyn Road Campus, Kingston upon Thames, London KT1 2EE, UK; (F.B.N.); (D.M.)
- Richards Medical Research Laboratories, Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Floor 7, 3700 Hamilton Walk, Philadelphia, PA 19104, USA
| |
Collapse
|
12
|
Yun J, Yun S, Park JE, Cheong EN, Park SY, Kim N, Kim HS. Deep Learning of Time-Signal Intensity Curves from Dynamic Susceptibility Contrast Imaging Enables Tissue Labeling and Prediction of Survival in Glioblastoma. AJNR Am J Neuroradiol 2023; 44:543-552. [PMID: 37105676 PMCID: PMC10171378 DOI: 10.3174/ajnr.a7853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 03/21/2023] [Indexed: 04/29/2023]
Abstract
BACKGROUND AND PURPOSE An autoencoder can learn representative time-signal intensity patterns to provide tissue heterogeneity measures using dynamic susceptibility contrast MR imaging. The aim of this study was to investigate whether such an autoencoder-based pattern analysis could provide interpretable tissue labeling and prognostic value in isocitrate dehydrogenase (IDH) wild-type glioblastoma. MATERIALS AND METHODS Preoperative dynamic susceptibility contrast MR images were obtained from 272 patients with IDH wild-type glioblastoma (training and validation, 183 and 89 patients, respectively). The autoencoder was applied to the dynamic susceptibility contrast MR imaging time-signal intensity curves of tumor and peritumoral areas. Representative perfusion patterns were defined by voxelwise K-means clustering using autoencoder latent features. Perfusion patterns were labeled by comparing parameters with anatomic reference tissues for baseline, signal drop, and percentage recovery. In the validation set (n = 89), a survival model was created from representative patterns and clinical predictors using Cox proportional hazard regression analysis, and its performance was calculated using the Harrell C-index. RESULTS Eighty-nine patients were enrolled. Five representative perfusion patterns were used to characterize tissues as high angiogenic tumor, low angiogenic/cellular tumor, perinecrotic lesion, infiltrated edema, and vasogenic edema. Of these, the low angiogenic/cellular tumor (hazard ratio, 2.18; P = .047) and infiltrated edema patterns (hazard ratio, 1.88; P = .009) in peritumoral areas showed significant prognostic value. The combined perfusion patterns and clinical predictors (C-index, 0.72) improved prognostication when added to clinical predictors (C-index, 0.55). CONCLUSIONS The autoencoder perfusion pattern analysis enabled tissue characterization of peritumoral areas, providing heterogeneity and dynamic information that may provide useful prognostic information in IDH wild-type glioblastoma.
Collapse
Affiliation(s)
- J Yun
- From the Departments of Convergence Medicine (J.Y., N.K.)
- Radiology and Research Institute of Radiology (J.Y., J.E.P., N.K., H.S.K.), Asan Medical Center
| | - S Yun
- Department of Radiology (S.Y.), Busan Paik Hospital, Inje University College of Medicine, Busan, Korea
| | - J E Park
- Radiology and Research Institute of Radiology (J.Y., J.E.P., N.K., H.S.K.), Asan Medical Center
| | - E-N Cheong
- Medical Science and Asan Medical Institute of Convergence Science and Technology (E.-N.C.), University of Ulsan College of Medicine, Seoul, Korea
| | - S Y Park
- Department of Statistics and Data Science (S.Y.P.), Korea National Open University, Seoul, Korea
| | - N Kim
- From the Departments of Convergence Medicine (J.Y., N.K.)
- Radiology and Research Institute of Radiology (J.Y., J.E.P., N.K., H.S.K.), Asan Medical Center
| | - H S Kim
- Radiology and Research Institute of Radiology (J.Y., J.E.P., N.K., H.S.K.), Asan Medical Center
| |
Collapse
|
13
|
Forecasting Molecular Features in IDH-Wildtype Gliomas: The State of the Art of Radiomics Applied to Neurosurgery. Cancers (Basel) 2023; 15:cancers15030940. [PMID: 36765898 PMCID: PMC9913449 DOI: 10.3390/cancers15030940] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 01/24/2023] [Accepted: 01/29/2023] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND The fifth edition of the WHO Classification of Tumors of the Central Nervous System (CNS), published in 2021, marks a step forward the future diagnostic approach to these neoplasms. Alongside this, radiomics has experienced rapid evolution over the last several years, allowing us to correlate tumor imaging heterogeneity with a wide range of tumor molecular and subcellular features. Radiomics is a translational field focused on decoding conventional imaging data to extrapolate the molecular and prognostic features of tumors such as gliomas. We herein analyze the state-of-the-art of radiomics applied to glioblastoma, with the goal to estimate its current clinical impact and potential perspectives in relation to well-rounded patient management, including the end-of-life stage. METHODS A literature review was performed on the PubMed, MEDLINE and Scopus databases using the following search items: "radiomics and glioma", "radiomics and glioblastoma", "radiomics and glioma and IDH", "radiomics and glioma and TERT promoter", "radiomics and glioma and EGFR", "radiomics and glioma and chromosome". RESULTS A total of 719 articles were screened. Further quantitative and qualitative analysis allowed us to finally include 11 papers. This analysis shows that radiomics is rapidly evolving towards a reliable tool. CONCLUSIONS Further studies are necessary to adjust radiomics' potential to the newest molecular requirements pointed out by the 2021 WHO classification of CNS tumors. At a glance, its application in the clinical routine could be beneficial to achieve a timely diagnosis, especially for those patients not eligible for surgery and/or adjuvant therapies but still deserving palliative and supportive care.
Collapse
|
14
|
Glioma radiogenomics and artificial intelligence: road to precision cancer medicine. Clin Radiol 2023; 78:137-149. [PMID: 36241568 DOI: 10.1016/j.crad.2022.08.138] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 08/19/2022] [Indexed: 01/18/2023]
Abstract
Radiogenomics refers to the study of the relationship between imaging phenotypes and gene expression patterns/molecular characteristics, which might allow improved diagnosis, decision-making, and predicting patient outcomes in the context of multiple diseases. Central nervous system (CNS) tumours contribute to significant cancer-related mortality in the present age. Although historically CNS neoplasms were classified and graded based on microscopic appearance, there was discordance between two histologically similar tumours that showed varying prognosis and behaviour, attributable to their molecular signatures. These led to the incorporation of molecular markers in the classification of CNS neoplasms. Meanwhile, advancements in imaging technology such as diffusion-based imaging (including tractography), perfusion, and spectroscopy in addition to the conventional imaging of glial neoplasms, have opened an avenue for radiogenomics. This review touches upon the schema of the current classification of gliomas, concepts behind molecular markers, and parameters that are used in radiogenomics to characterise gliomas and the role of artificial intelligence for the same. Further, the role of radiomics in the grading of brain tumours, prediction of treatment response and prognosis has been discussed. Use of automated and semi-automated tumour segmentation for radiotherapy planning and follow-up has also been discussed briefly.
Collapse
|
15
|
Cellina M, Cè M, Khenkina N, Sinichich P, Cervelli M, Poggi V, Boemi S, Ierardi AM, Carrafiello G. Artificial Intellgence in the Era of Precision Oncological Imaging. Technol Cancer Res Treat 2022; 21:15330338221141793. [PMID: 36426565 PMCID: PMC9703524 DOI: 10.1177/15330338221141793] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Rapid-paced development and adaptability of artificial intelligence algorithms have secured their almost ubiquitous presence in the field of oncological imaging. Artificial intelligence models have been created for a variety of tasks, including risk stratification, automated detection, and segmentation of lesions, characterization, grading and staging, prediction of prognosis, and treatment response. Soon, artificial intelligence could become an essential part of every step of oncological workup and patient management. Integration of neural networks and deep learning into radiological artificial intelligence algorithms allow for extrapolating imaging features otherwise inaccessible to human operators and pave the way to truly personalized management of oncological patients.Although a significant proportion of currently available artificial intelligence solutions belong to basic and translational cancer imaging research, their progressive transfer to clinical routine is imminent, contributing to the development of a personalized approach in oncology. We thereby review the main applications of artificial intelligence in oncological imaging, describe the example of their successful integration into research and clinical practice, and highlight the challenges and future perspectives that will shape the field of oncological radiology.
Collapse
Affiliation(s)
- Michaela Cellina
- Radiology Department, Fatebenefratelli Hospital, Milano, Italy,Michaela Cellina, MD, Radiology Department, Fatebenefratelli Hospital, ASST Fatebenefratelli Sacco, Milano, Piazza Principessa Clotilde 3, 20121, Milano, Italy.
| | - Maurizio Cè
- Postgraduate School in Radiodiagnostics, Università degli Studi di Milano, Milan, Italy
| | - Natallia Khenkina
- Postgraduate School in Radiodiagnostics, Università degli Studi di Milano, Milan, Italy
| | - Polina Sinichich
- Postgraduate School in Radiodiagnostics, Università degli Studi di Milano, Milan, Italy
| | - Marco Cervelli
- Postgraduate School in Radiodiagnostics, Università degli Studi di Milano, Milan, Italy
| | - Vittoria Poggi
- Postgraduate School in Radiodiagnostics, Università degli Studi di Milano, Milan, Italy
| | - Sara Boemi
- Postgraduate School in Radiodiagnostics, Università degli Studi di Milano, Milan, Italy
| | | | - Gianpaolo Carrafiello
- Postgraduate School in Radiodiagnostics, Università degli Studi di Milano, Milan, Italy,Radiology Department, Fondazione IRCCS Cà Granda, Milan, Italy
| |
Collapse
|
16
|
Pati S, Baid U, Edwards B, Sheller MJ, Foley P, Reina GA, Thakur S, Sako C, Bilello M, Davatzikos C, Martin J, Shah P, Menze B, Bakas S. The federated tumor segmentation (FeTS) tool: an open-source solution to further solid tumor research. Phys Med Biol 2022; 67:10.1088/1361-6560/ac9449. [PMID: 36137534 PMCID: PMC9592188 DOI: 10.1088/1361-6560/ac9449] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 09/22/2022] [Indexed: 11/11/2022]
Abstract
Objective.De-centralized data analysis becomes an increasingly preferred option in the healthcare domain, as it alleviates the need for sharing primary patient data across collaborating institutions. This highlights the need for consistent harmonized data curation, pre-processing, and identification of regions of interest based on uniform criteria.Approach.Towards this end, this manuscript describes theFederatedTumorSegmentation (FeTS) tool, in terms of software architecture and functionality.Main results.The primary aim of the FeTS tool is to facilitate this harmonized processing and the generation of gold standard reference labels for tumor sub-compartments on brain magnetic resonance imaging, and further enable federated training of a tumor sub-compartment delineation model across numerous sites distributed across the globe, without the need to share patient data.Significance.Building upon existing open-source tools such as the Insight Toolkit and Qt, the FeTS tool is designed to enable training deep learning models targeting tumor delineation in either centralized or federated settings. The target audience of the FeTS tool is primarily the computational researcher interested in developing federated learning models, and interested in joining a global federation towards this effort. The tool is open sourced athttps://github.com/FETS-AI/Front-End.
Collapse
Affiliation(s)
- Sarthak Pati
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Informatics, Technical University of Munich, Munich, Germany
| | - Ujjwal Baid
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | | | | | - Siddhesh Thakur
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Chiharu Sako
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michel Bilello
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christos Davatzikos
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | - Bjoern Menze
- Department of Informatics, Technical University of Munich, Munich, Germany
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
| | - Spyridon Bakas
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
17
|
Wu J, Mayer AT, Li R. Integrated imaging and molecular analysis to decipher tumor microenvironment in the era of immunotherapy. Semin Cancer Biol 2022; 84:310-328. [PMID: 33290844 PMCID: PMC8319834 DOI: 10.1016/j.semcancer.2020.12.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 11/29/2020] [Accepted: 12/02/2020] [Indexed: 02/07/2023]
Abstract
Radiological imaging is an integral component of cancer care, including diagnosis, staging, and treatment response monitoring. It contains rich information about tumor phenotypes that are governed not only by cancer cellintrinsic biological processes but also by the tumor microenvironment, such as the composition and function of tumor-infiltrating immune cells. By analyzing the radiological scans using a quantitative radiomics approach, robust relations between specific imaging and molecular phenotypes can be established. Indeed, a number of studies have demonstrated the feasibility of radiogenomics for predicting intrinsic molecular subtypes and gene expression signatures in breast cancer based on MRI. In parallel, promising results have been shown for inferring the amount of tumor-infiltrating lymphocytes, a key factor for the efficacy of cancer immunotherapy, from standard-of-care radiological images. Compared with the biopsy-based approach, radiogenomics offers a unique avenue to profile the molecular makeup of the tumor and immune microenvironment as well as its evolution in a noninvasive and holistic manner through longitudinal imaging scans. Here, we provide a systematic review of the state of the art radiogenomics studies in the era of immunotherapy and discuss emerging paradigms and opportunities in AI and deep learning approaches. These technical advances are expected to transform the radiogenomics field, leading to the discovery of reliable imaging biomarkers. This will pave the way for their clinical translation to guide precision cancer therapy.
Collapse
Affiliation(s)
- Jia Wu
- Department of Imaging Physics, MD Anderson Cancer Center, Texas, 77030, USA; Department of Thoracic/Head & Neck Medical Oncology, MD Anderson Cancer Center, Texas, 77030, USA.
| | - Aaron T Mayer
- Department of Bioengineering, Stanford University, Stanford, California, 94305, USA; Department of Radiology, Stanford University, Stanford, California, 94305, USA; Molecular Imaging Program at Stanford, Stanford University, Stanford, California, 94305, USA; BioX Program at Stanford, Stanford University, Stanford, California, 94305, USA
| | - Ruijiang Li
- Department of Radiation Oncology, Stanford University, Stanford, California, 94305, USA
| |
Collapse
|
18
|
Spatial heterogeneity of edema region uncovers survival-relevant habitat of Glioblastoma. Eur J Radiol 2022; 154:110423. [DOI: 10.1016/j.ejrad.2022.110423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 05/16/2022] [Accepted: 06/20/2022] [Indexed: 11/18/2022]
|
19
|
Sansone G, Vivori N, Vivori C, Di Stefano AL, Picca A. Basic premises: searching for new targets and strategies in diffuse gliomas. Clin Transl Imaging 2022. [DOI: 10.1007/s40336-022-00507-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
20
|
Bakas S, Sako C, Akbari H, Bilello M, Sotiras A, Shukla G, Rudie JD, Santamaría NF, Kazerooni AF, Pati S, Rathore S, Mamourian E, Ha SM, Parker W, Doshi J, Baid U, Bergman M, Binder ZA, Verma R, Lustig RA, Desai AS, Bagley SJ, Mourelatos Z, Morrissette J, Watt CD, Brem S, Wolf RL, Melhem ER, Nasrallah MP, Mohan S, O'Rourke DM, Davatzikos C. The University of Pennsylvania glioblastoma (UPenn-GBM) cohort: advanced MRI, clinical, genomics, & radiomics. Sci Data 2022; 9:453. [PMID: 35906241 PMCID: PMC9338035 DOI: 10.1038/s41597-022-01560-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 07/12/2022] [Indexed: 02/05/2023] Open
Abstract
Glioblastoma is the most common aggressive adult brain tumor. Numerous studies have reported results from either private institutional data or publicly available datasets. However, current public datasets are limited in terms of: a) number of subjects, b) lack of consistent acquisition protocol, c) data quality, or d) accompanying clinical, demographic, and molecular information. Toward alleviating these limitations, we contribute the "University of Pennsylvania Glioblastoma Imaging, Genomics, and Radiomics" (UPenn-GBM) dataset, which describes the currently largest publicly available comprehensive collection of 630 patients diagnosed with de novo glioblastoma. The UPenn-GBM dataset includes (a) advanced multi-parametric magnetic resonance imaging scans acquired during routine clinical practice, at the University of Pennsylvania Health System, (b) accompanying clinical, demographic, and molecular information, (d) perfusion and diffusion derivative volumes, (e) computationally-derived and manually-revised expert annotations of tumor sub-regions, as well as (f) quantitative imaging (also known as radiomic) features corresponding to each of these regions. This collection describes our contribution towards repeatable, reproducible, and comparative quantitative studies leading to new predictive, prognostic, and diagnostic assessments.
Collapse
Affiliation(s)
- Spyridon Bakas
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Chiharu Sako
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hamed Akbari
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michel Bilello
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Aristeidis Sotiras
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology and Institute for Informatics, Washington University, School of Medicine, St. Louis, MO, USA
| | - Gaurav Shukla
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiation Oncology, Christiana Care Health System, Philadelphia, PA, USA
| | - Jeffrey D Rudie
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology & Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - Natali Flores Santamaría
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Anahita Fathi Kazerooni
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sarthak Pati
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Saima Rathore
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth Mamourian
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sung Min Ha
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology and Institute for Informatics, Washington University, School of Medicine, St. Louis, MO, USA
| | - William Parker
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jimit Doshi
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ujjwal Baid
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mark Bergman
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
| | - Zev A Binder
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ragini Verma
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Robert A Lustig
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Arati S Desai
- Division of Hematology Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Stephen J Bagley
- Division of Hematology Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zissimos Mourelatos
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jennifer Morrissette
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christopher D Watt
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Steven Brem
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ronald L Wolf
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elias R Melhem
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - MacLean P Nasrallah
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Suyash Mohan
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Donald M O'Rourke
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christos Davatzikos
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA.
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
21
|
Sayah A, Bencheqroun C, Bhuvaneshwar K, Belouali A, Bakas S, Sako C, Davatzikos C, Alaoui A, Madhavan S, Gusev Y. Enhancing the REMBRANDT MRI collection with expert segmentation labels and quantitative radiomic features. Sci Data 2022; 9:338. [PMID: 35701399 PMCID: PMC9198015 DOI: 10.1038/s41597-022-01415-1] [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: 10/12/2021] [Accepted: 05/24/2022] [Indexed: 01/26/2023] Open
Abstract
Malignancy of the brain and CNS is unfortunately a common diagnosis. A large subset of these lesions tends to be high grade tumors which portend poor prognoses and low survival rates, and are estimated to be the tenth leading cause of death worldwide. The complex nature of the brain tissue environment in which these lesions arise offers a rich opportunity for translational research. Magnetic Resonance Imaging (MRI) can provide a comprehensive view of the abnormal regions in the brain, therefore, its applications in the translational brain cancer research is considered essential for the diagnosis and monitoring of disease. Recent years has seen rapid growth in the field of radiogenomics, especially in cancer, and scientists have been able to successfully integrate the quantitative data extracted from medical images (also known as radiomics) with genomics to answer new and clinically relevant questions. In this paper, we took raw MRI scans from the REMBRANDT data collection from public domain, and performed volumetric segmentation to identify subregions of the brain. Radiomic features were then extracted to represent the MRIs in a quantitative yet summarized format. This resulting dataset now enables further biomedical and integrative data analysis, and is being made public via the NeuroImaging Tools & Resources Collaboratory (NITRC) repository ( https://www.nitrc.org/projects/rembrandt_brain/ ).
Collapse
Affiliation(s)
- Anousheh Sayah
- Medstar Georgetown University Hospital, Washington, DC, USA
| | - Camelia Bencheqroun
- Innovation Center for Biomedical Informatics (ICBI), Georgetown University, Washington, DC, USA
| | - Krithika Bhuvaneshwar
- Innovation Center for Biomedical Informatics (ICBI), Georgetown University, Washington, DC, USA.
| | - Anas Belouali
- Innovation Center for Biomedical Informatics (ICBI), Georgetown University, Washington, DC, USA
| | - Spyridon Bakas
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
| | - Chiharu Sako
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
| | - Christos Davatzikos
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
| | - Adil Alaoui
- Innovation Center for Biomedical Informatics (ICBI), Georgetown University, Washington, DC, USA
| | - Subha Madhavan
- Innovation Center for Biomedical Informatics (ICBI), Georgetown University, Washington, DC, USA
| | - Yuriy Gusev
- Innovation Center for Biomedical Informatics (ICBI), Georgetown University, Washington, DC, USA.
| |
Collapse
|
22
|
Non-invasive molecular diagnosis in gliomas with advanced imaging. Clin Transl Imaging 2022. [DOI: 10.1007/s40336-022-00501-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
23
|
Corr F, Grimm D, Saß B, Pojskić M, Bartsch JW, Carl B, Nimsky C, Bopp MHA. Radiogenomic Predictors of Recurrence in Glioblastoma—A Systematic Review. J Pers Med 2022; 12:jpm12030402. [PMID: 35330402 PMCID: PMC8952807 DOI: 10.3390/jpm12030402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 02/23/2022] [Accepted: 03/01/2022] [Indexed: 12/10/2022] Open
Abstract
Glioblastoma, as the most aggressive brain tumor, is associated with a poor prognosis and outcome. To optimize prognosis and clinical therapy decisions, there is an urgent need to stratify patients with increased risk for recurrent tumors and low therapeutic success to optimize individual treatment. Radiogenomics establishes a link between radiological and pathological information. This review provides a state-of-the-art picture illustrating the latest developments in the use of radiogenomic markers regarding prognosis and their potential for monitoring recurrence. Databases PubMed, Google Scholar, and Cochrane Library were searched. Inclusion criteria were defined as diagnosis of glioblastoma with histopathological and radiological follow-up. Out of 321 reviewed articles, 43 articles met these inclusion criteria. Included studies were analyzed for the frequency of radiological and molecular tumor markers whereby radiogenomic associations were analyzed. Six main associations were described: radiogenomic prognosis, MGMT status, IDH, EGFR status, molecular subgroups, and tumor location. Prospective studies analyzing prognostic features of glioblastoma together with radiological features are lacking. By reviewing the progress in the development of radiogenomic markers, we provide insights into the potential efficacy of such an approach for clinical routine use eventually enabling early identification of glioblastoma recurrence and therefore supporting a further personalized monitoring and treatment strategy.
Collapse
Affiliation(s)
- Felix Corr
- Department of Neurosurgery, University of Marburg, Baldingerstrasse, 35043 Marburg, Germany; (D.G.); (B.S.); (M.P.); (J.W.B.); (B.C.); (C.N.); (M.H.A.B.)
- EDU Institute of Higher Education, Villa Bighi, Chaplain’s House, KKR 1320 Kalkara, Malta
- Correspondence:
| | - Dustin Grimm
- Department of Neurosurgery, University of Marburg, Baldingerstrasse, 35043 Marburg, Germany; (D.G.); (B.S.); (M.P.); (J.W.B.); (B.C.); (C.N.); (M.H.A.B.)
- EDU Institute of Higher Education, Villa Bighi, Chaplain’s House, KKR 1320 Kalkara, Malta
| | - Benjamin Saß
- Department of Neurosurgery, University of Marburg, Baldingerstrasse, 35043 Marburg, Germany; (D.G.); (B.S.); (M.P.); (J.W.B.); (B.C.); (C.N.); (M.H.A.B.)
| | - Mirza Pojskić
- Department of Neurosurgery, University of Marburg, Baldingerstrasse, 35043 Marburg, Germany; (D.G.); (B.S.); (M.P.); (J.W.B.); (B.C.); (C.N.); (M.H.A.B.)
| | - Jörg W. Bartsch
- Department of Neurosurgery, University of Marburg, Baldingerstrasse, 35043 Marburg, Germany; (D.G.); (B.S.); (M.P.); (J.W.B.); (B.C.); (C.N.); (M.H.A.B.)
- Center for Mind, Brain and Behavior (CMBB), 35043 Marburg, Germany
| | - Barbara Carl
- Department of Neurosurgery, University of Marburg, Baldingerstrasse, 35043 Marburg, Germany; (D.G.); (B.S.); (M.P.); (J.W.B.); (B.C.); (C.N.); (M.H.A.B.)
- Department of Neurosurgery, Helios Dr. Horst Schmidt Kliniken, Ludwig-Erhard-Strasse 100, 65199 Wiesbaden, Germany
| | - Christopher Nimsky
- Department of Neurosurgery, University of Marburg, Baldingerstrasse, 35043 Marburg, Germany; (D.G.); (B.S.); (M.P.); (J.W.B.); (B.C.); (C.N.); (M.H.A.B.)
- Center for Mind, Brain and Behavior (CMBB), 35043 Marburg, Germany
| | - Miriam H. A. Bopp
- Department of Neurosurgery, University of Marburg, Baldingerstrasse, 35043 Marburg, Germany; (D.G.); (B.S.); (M.P.); (J.W.B.); (B.C.); (C.N.); (M.H.A.B.)
- Center for Mind, Brain and Behavior (CMBB), 35043 Marburg, Germany
| |
Collapse
|
24
|
Güley O, Pati S, Bakas S. Classification of Infection and Ischemia in Diabetic Foot Ulcers Using VGG Architectures. DIABETIC FOOT ULCERS GRAND CHALLENGE : SECOND CHALLENGE, DFUC 2021, HELD IN CONJUNCTION WITH MICCAI 2021, STRASBOURG, FRANCE, SEPTEMBER 27, 2021 : PROCEEDINGS. DFUC (CONFERENCE) (2ND : 2021 : ONLINE) 2022; 13183:76-89. [PMID: 35465060 PMCID: PMC9026672 DOI: 10.1007/978-3-030-94907-5_6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Diabetic foot ulceration (DFU) is a serious complication of diabetes, and a major challenge for healthcare systems around the world. Further infection and ischemia in DFU can significantly prolong treatment and often result in limb amputation, with more severe cases resulting in terminal illness. Thus, early identification and regular monitoring is necessary to improve care, and reduce the burden on healthcare systems. With that in mind, this study attempts to address the problem of infection and ischemia classification in diabetic food ulcers, in four distinct classes. We have evaluated a series of VGG architectures with different layers, following numerous training strategies, including k-fold cross validation, data pre-processing options, augmentation techniques, and weighted loss calculations. In favor of transparency and reproducibility, we make all the implementations available through the Generally Nuanced Deep Learning Framework (GaNDLF, github.com/CBICA/GaNDLF. Our best model was evaluated during the DFU Challenge 2021, and was ranked 2nd, 5th, and 7th based on the macro-averaged AUC (area under the curve), macro-averaged F1 score, and macro-averaged recall metrics, respectively. Our findings support that current state-of-the-art architectures provide good results for the DFU image classification task, and further experimentation is required to study the effects of pre-processing and augmentation strategies.
Collapse
Affiliation(s)
- Orhun Güley
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Informatics, Technical University of Munich, Munich, Germany
| | - Sarthak Pati
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Informatics, Technical University of Munich, Munich, Germany
| | - Spyridon Bakas
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
25
|
Fathi Kazerooni A, Bagley SJ, Akbari H, Saxena S, Bagheri S, Guo J, Chawla S, Nabavizadeh A, Mohan S, Bakas S, Davatzikos C, Nasrallah MP. Applications of Radiomics and Radiogenomics in High-Grade Gliomas in the Era of Precision Medicine. Cancers (Basel) 2021; 13:cancers13235921. [PMID: 34885031 PMCID: PMC8656630 DOI: 10.3390/cancers13235921] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/19/2021] [Accepted: 11/22/2021] [Indexed: 12/22/2022] Open
Abstract
Simple Summary Radiomics and radiogenomics offer new insight into high-grade glioma biology, as well as into glioma behavior in response to standard therapies. In this article, we provide neuro-oncology, neuropathology, and computational perspectives on the role of radiomics in providing more accurate diagnoses, prognostication, and surveillance of patients with high-grade glioma, and on the potential application of radiomics in clinical practice, with the overarching goal of advancing precision medicine for optimal patient care. Abstract Machine learning (ML) integrated with medical imaging has introduced new perspectives in precision diagnostics of high-grade gliomas, through radiomics and radiogenomics. This has raised hopes for characterizing noninvasive and in vivo biomarkers for prediction of patient survival, tumor recurrence, and genomics and therefore encouraging treatments tailored to individualized needs. Characterization of tumor infiltration based on pre-operative multi-parametric magnetic resonance imaging (MP-MRI) scans may allow prediction of the loci of future tumor recurrence and thereby aid in planning the course of treatment for the patients, such as optimizing the extent of resection and the dose and target area of radiation. Imaging signatures of tumor genomics can help in identifying the patients who benefit from certain targeted therapies. Specifying molecular properties of gliomas and prediction of their changes over time and with treatment would allow optimization of treatment. In this article, we provide neuro-oncology, neuropathology, and computational perspectives on the promise of radiomics and radiogenomics for allowing personalized treatments of patients with gliomas and discuss the challenges and limitations of these methods in multi-institutional clinical trials and suggestions to mitigate the issues and the future directions.
Collapse
Affiliation(s)
- Anahita Fathi Kazerooni
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA 19104, USA; (A.F.K.); (H.A.); (S.S.); (J.G.); (A.N.); (S.M.); (S.B.); (C.D.)
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.B.); (S.C.)
| | - Stephen J. Bagley
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hamed Akbari
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA 19104, USA; (A.F.K.); (H.A.); (S.S.); (J.G.); (A.N.); (S.M.); (S.B.); (C.D.)
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.B.); (S.C.)
| | - Sanjay Saxena
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA 19104, USA; (A.F.K.); (H.A.); (S.S.); (J.G.); (A.N.); (S.M.); (S.B.); (C.D.)
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.B.); (S.C.)
| | - Sina Bagheri
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.B.); (S.C.)
| | - Jun Guo
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA 19104, USA; (A.F.K.); (H.A.); (S.S.); (J.G.); (A.N.); (S.M.); (S.B.); (C.D.)
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.B.); (S.C.)
| | - Sanjeev Chawla
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.B.); (S.C.)
| | - Ali Nabavizadeh
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA 19104, USA; (A.F.K.); (H.A.); (S.S.); (J.G.); (A.N.); (S.M.); (S.B.); (C.D.)
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.B.); (S.C.)
| | - Suyash Mohan
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA 19104, USA; (A.F.K.); (H.A.); (S.S.); (J.G.); (A.N.); (S.M.); (S.B.); (C.D.)
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.B.); (S.C.)
| | - Spyridon Bakas
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA 19104, USA; (A.F.K.); (H.A.); (S.S.); (J.G.); (A.N.); (S.M.); (S.B.); (C.D.)
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.B.); (S.C.)
- Department of Pathology & Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Christos Davatzikos
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA 19104, USA; (A.F.K.); (H.A.); (S.S.); (J.G.); (A.N.); (S.M.); (S.B.); (C.D.)
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.B.); (S.C.)
| | - MacLean P. Nasrallah
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pathology & Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Correspondence:
| |
Collapse
|
26
|
Jaberipour M, Soliman H, Sahgal A, Sadeghi-Naini A. A priori prediction of local failure in brain metastasis after hypo-fractionated stereotactic radiotherapy using quantitative MRI and machine learning. Sci Rep 2021; 11:21620. [PMID: 34732781 PMCID: PMC8566533 DOI: 10.1038/s41598-021-01024-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 10/21/2021] [Indexed: 12/14/2022] Open
Abstract
This study investigated the effectiveness of pre-treatment quantitative MRI and clinical features along with machine learning techniques to predict local failure in patients with brain metastasis treated with hypo-fractionated stereotactic radiation therapy (SRT). The predictive models were developed using the data from 100 patients (141 lesions) and evaluated on an independent test set with data from 20 patients (30 lesions). Quantitative MRI radiomic features were derived from the treatment-planning contrast-enhanced T1w and T2-FLAIR images. A multi-phase feature reduction and selection procedure was applied to construct an optimal quantitative MRI biomarker for predicting therapy outcome. The performance of standard clinical features in therapy outcome prediction was evaluated using a similar procedure. Survival analyses were conducted to compare the long-term outcome of the two patient cohorts (local control/failure) identified based on prediction at pre-treatment, and standard clinical criteria at last patient follow-up after SRT. The developed quantitative MRI biomarker consists of four features with two features quantifying heterogeneity in the edema region, one feature characterizing intra-tumour heterogeneity, and one feature describing tumour morphology. The predictive models with the radiomic and clinical feature sets yielded an AUC of 0.87 and 0.62, respectively on the independent test set. Incorporating radiomic features into the clinical predictive model improved the AUC of the model by up to 16%, relatively. A statistically significant difference was observed in survival of the two patient cohorts identified at pre-treatment using the radiomics-based predictive model, and at post-treatment using the the RANO-BM criteria. Results of this study revealed a good potential for quantitative MRI radiomic features at pre-treatment in predicting local failure in relatively large brain metastases undergoing SRT, and is a step forward towards a precision oncology paradigm for brain metastasis.
Collapse
Affiliation(s)
- Majid Jaberipour
- grid.21100.320000 0004 1936 9430Department of Electrical Engineering and Computer Science, Lassonde School of Engineering, York University, Toronto, ON Canada ,grid.413104.30000 0000 9743 1587Physical Sciences Platform, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON Canada
| | - Hany Soliman
- grid.413104.30000 0000 9743 1587Physical Sciences Platform, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON Canada ,grid.413104.30000 0000 9743 1587Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON Canada ,grid.17063.330000 0001 2157 2938Department of Radiation Oncology, University of Toronto, Toronto, ON Canada
| | - Arjun Sahgal
- grid.413104.30000 0000 9743 1587Physical Sciences Platform, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON Canada ,grid.413104.30000 0000 9743 1587Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON Canada ,grid.17063.330000 0001 2157 2938Department of Radiation Oncology, University of Toronto, Toronto, ON Canada
| | - Ali Sadeghi-Naini
- grid.21100.320000 0004 1936 9430Department of Electrical Engineering and Computer Science, Lassonde School of Engineering, York University, Toronto, ON Canada ,grid.413104.30000 0000 9743 1587Physical Sciences Platform, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON Canada ,grid.413104.30000 0000 9743 1587Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON Canada ,grid.17063.330000 0001 2157 2938Department of Medical Biophysics, University of Toronto, Toronto, ON Canada
| |
Collapse
|
27
|
Zhang X, Katsakhyan L, LiVolsi VA, Roth JJ, Rassekh CH, Bagley SJ, Nasrallah MP. TP53 Mutation and Extraneural Metastasis of Glioblastoma: Insights From an Institutional Experience and Comprehensive Literature Review. Am J Surg Pathol 2021; 45:1516-1526. [PMID: 34366423 DOI: 10.1097/pas.0000000000001762] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Extraneural metastases of glioblastoma (GBM), although rare, are becoming an increasingly recognized occurrence. Currently, the biological mechanism underlying this rare occurrence is not understood. To explore the potential genomic drivers of extraneural metastasis in GBM, we present the molecular features of 4 extraneural metastatic GBMs, along with a comprehensive review and analysis of previously reported cases that had available molecular characterization. In addition to our 4 cases, 42 patients from 35 publications are reviewed. To compare the molecular profiles between GBM cases with extraneural metastasis and the general GBM population, genomic data from GBM samples in The Cancer Genome Atlas (TCGA) database were also analyzed. We found that 64.5% (20/31) of the cases with extraneural metastasis that were tested for TP53 changes had at least 1 TP53 pathogenic variant detected in either 1 or both primary and metastatic tumors. In contrast, TP53 mutation was significantly less frequent in the unselected GBM from TCGA (22.6%, 56/248) (P=0.000). In addition, O-6-methylguanine-DNA methyltransferase (MGMT) promoter methylation was more common in unselected TCGA GBM cases (48.6%, 170/350) than in cases with extraneural metastasis (31.8%, 7/22), although not statistically significant. Although isocitrate dehydrogenase (IDH) mutation is a rare occurrence in high-grade astrocytomas, IDH-mutant grade 4 astrocytomas are at least as likely to metastasize as IDH wild-type GBMs; 3 metastatic cases definitively harbored an IDH1 (p.R132H) mutation in our analysis. Our findings not only provide potential biomarkers for earlier screening of extraneural metastasis, but could also suggest clues to understanding biological mechanisms underlying GBM metastasis, and for the development of therapeutic modalities.
Collapse
Affiliation(s)
| | | | | | | | | | - Stephen J Bagley
- Hematology Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA
| | | |
Collapse
|
28
|
Akbari H, Kazerooni AF, Ware JB, Mamourian E, Anderson H, Guiry S, Sako C, Raymond C, Yao J, Brem S, O'Rourke DM, Desai AS, Bagley SJ, Ellingson BM, Davatzikos C, Nabavizadeh A. Quantification of tumor microenvironment acidity in glioblastoma using principal component analysis of dynamic susceptibility contrast enhanced MR imaging. Sci Rep 2021; 11:15011. [PMID: 34294864 PMCID: PMC8298590 DOI: 10.1038/s41598-021-94560-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 06/28/2021] [Indexed: 11/22/2022] Open
Abstract
Glioblastoma (GBM) has high metabolic demands, which can lead to acidification of the tumor microenvironment. We hypothesize that a machine learning model built on temporal principal component analysis (PCA) of dynamic susceptibility contrast-enhanced (DSC) perfusion MRI can be used to estimate tumor acidity in GBM, as estimated by pH-sensitive amine chemical exchange saturation transfer echo-planar imaging (CEST-EPI). We analyzed 78 MRI scans in 32 treatment naïve and post-treatment GBM patients. All patients were imaged with DSC-MRI, and pH-weighting that was quantified from CEST-EPI estimation of the magnetization transfer ratio asymmetry (MTRasym) at 3 ppm. Enhancing tumor (ET), non-enhancing core (NC), and peritumoral T2 hyperintensity (namely, edema, ED) were used to extract principal components (PCs) and to build support vector machines regression (SVR) models to predict MTRasym values using PCs. Our predicted map correlated with MTRasym values with Spearman's r equal to 0.66, 0.47, 0.67, 0.71, in NC, ET, ED, and overall, respectively (p < 0.006). The results of this study demonstrates that PCA analysis of DSC imaging data can provide information about tumor pH in GBM patients, with the strongest association within the peritumoral regions.
Collapse
Affiliation(s)
- Hamed Akbari
- Department of Radiology, Perelman School of Medicine, Hospital of University of Pennsylvania, University of Pennsylvania, Philadelphia, PA, USA
- Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Anahita Fathi Kazerooni
- Department of Radiology, Perelman School of Medicine, Hospital of University of Pennsylvania, University of Pennsylvania, Philadelphia, PA, USA
- Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jeffrey B Ware
- Department of Radiology, Perelman School of Medicine, Hospital of University of Pennsylvania, University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth Mamourian
- Department of Radiology, Perelman School of Medicine, Hospital of University of Pennsylvania, University of Pennsylvania, Philadelphia, PA, USA
- Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hannah Anderson
- Department of Radiology, Perelman School of Medicine, Hospital of University of Pennsylvania, University of Pennsylvania, Philadelphia, PA, USA
| | - Samantha Guiry
- Department of Radiology, Perelman School of Medicine, Hospital of University of Pennsylvania, University of Pennsylvania, Philadelphia, PA, USA
| | - Chiharu Sako
- Department of Radiology, Perelman School of Medicine, Hospital of University of Pennsylvania, University of Pennsylvania, Philadelphia, PA, USA
- Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Catalina Raymond
- UCLA Brain Tumor Imaging Laboratory, Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Jingwen Yao
- UCLA Brain Tumor Imaging Laboratory, Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Steven Brem
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Donald M O'Rourke
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Arati S Desai
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Stephen J Bagley
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Benjamin M Ellingson
- UCLA Brain Tumor Imaging Laboratory, Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Christos Davatzikos
- Department of Radiology, Perelman School of Medicine, Hospital of University of Pennsylvania, University of Pennsylvania, Philadelphia, PA, USA
- Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ali Nabavizadeh
- Department of Radiology, Perelman School of Medicine, Hospital of University of Pennsylvania, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
29
|
Identification of magnetic resonance imaging features for the prediction of molecular profiles of newly diagnosed glioblastoma. J Neurooncol 2021; 154:83-92. [PMID: 34191225 DOI: 10.1007/s11060-021-03801-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 06/25/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE We predicted molecular profiles in newly diagnosed glioblastoma patients using magnetic resonance (MR) imaging features and explored the associations between imaging features and major molecular alterations. METHODS This retrospective study included patients with newly diagnosed glioblastoma and available next-generation sequencing results. From preoperative MR imaging, Visually AcceSAble Rembrandt Images (VASARI) features, volumetric parameters, and apparent diffusion coefficient (ADC) values were obtained. First, univariate random forest was performed to identify gene abnormalities that could be predicted by imaging features with high accuracy and stability. Next, multivariate random forest was trained to predict the selected genes in the discovery cohort and was validated in the external cohort. Univariable logistic regression was performed to further explore the associations between imaging features and genes. RESULTS Univariate random forest identified nine genes predicted by imaging features, with high accuracy and stability. The multivariate random forest model showed excellent performance in predicting IDH and PTPN11 mutations in the discovery cohort, which were validated in the external validation cohorts (areas under the receiver operator characteristic curve [AUCs] of 0.855 for IDH and 0.88 for PTPN11). ATRX loss and EGFR mutation were predicted with AUCs of 0.753 and 0.739, respectively, whereas PTEN could not be reliably predicted. Based on univariable logistic regression analyses, IDH, ATRX, and TP53 were clustered according to their shared imaging features, whereas EGFR and CDKN2A/B were clustered in the opposite direction. CONCLUSIONS MR imaging features are related to specific molecular alterations and can be used to predict molecular profiles in patients with newly diagnosed glioblastoma.
Collapse
|
30
|
Singh A, Chitalia R, Kontos D. Radiogenomics in brain, breast, and lung cancer: opportunities and challenges. J Med Imaging (Bellingham) 2021; 8:031907. [PMID: 34164563 PMCID: PMC8212946 DOI: 10.1117/1.jmi.8.3.031907] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 06/04/2021] [Indexed: 01/06/2023] Open
Abstract
The field of radiogenomics largely focuses on developing imaging surrogates for genomic signatures and integrating imaging, genomic, and molecular data to develop combined personalized biomarkers for characterizing various diseases. Our study aims to highlight the current state-of-the-art and the role of radiogenomics in cancer research, focusing mainly on solid tumors, and is broadly divided into four sections. The first section reviews representative studies that establish the biologic basis of radiomic signatures using gene expression and molecular profiling information. The second section includes studies that aim to non-invasively predict molecular subtypes of tumors using radiomic signatures. The third section reviews studies that evaluate the potential to augment the performance of established prognostic signatures by combining complementary information encoded by radiomic and genomic signatures derived from cancer tumors. The fourth section includes studies that focus on ascertaining the biological significance of radiomic phenotypes. We conclude by discussing current challenges and opportunities in the field, such as the importance of coordination between imaging device manufacturers, regulatory organizations, health care providers, pharmaceutical companies, academic institutions, and physicians for the effective standardization of the results from radiogenomic signatures and for the potential use of these findings to improve precision care for cancer patients.
Collapse
Affiliation(s)
- Apurva Singh
- University of Pennsylvania, Department of Radiology, Philadelphia, Pennsylvania, United States
| | - Rhea Chitalia
- University of Pennsylvania, Department of Radiology, Philadelphia, Pennsylvania, United States
| | - Despina Kontos
- University of Pennsylvania, Department of Radiology, Philadelphia, Pennsylvania, United States
| |
Collapse
|
31
|
Differentiating Glioblastomas from Solitary Brain Metastases: An Update on the Current Literature of Advanced Imaging Modalities. Cancers (Basel) 2021; 13:cancers13122960. [PMID: 34199151 PMCID: PMC8231515 DOI: 10.3390/cancers13122960] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/09/2021] [Accepted: 06/09/2021] [Indexed: 12/12/2022] Open
Abstract
Differentiating between glioblastomas and solitary brain metastases proves to be a challenging diagnosis for neuroradiologists, as both present with imaging patterns consisting of peritumoral hyperintensities with similar intratumoral texture on traditional magnetic resonance imaging sequences. Early diagnosis is paramount, as each pathology has completely different methods of clinical assessment. In the past decade, recent developments in advanced imaging modalities enabled providers to acquire a more accurate diagnosis earlier in the patient's clinical assessment, thus optimizing clinical outcome. Dynamic susceptibility contrast has been optimized for detecting relative cerebral blood flow and relative cerebral blood volume. Diffusion tensor imaging can be used to detect changes in mean diffusivity. Neurite orientation dispersion and density imaging is an innovative modality detecting changes in intracellular volume fraction, isotropic volume fraction, and extracellular volume fraction. Magnetic resonance spectroscopy is able to assist by providing a metabolic descriptor while detecting variable ratios of choline/N-acetylaspartate, choline/creatine, and N-acetylaspartate/creatine. Finally, radiomics and machine learning algorithms have been devised to assist in improving diagnostic accuracy while often utilizing more than one advanced imaging protocol per patient. In this review, we provide an update on all the current evidence regarding the identification and differentiation of glioblastomas from solitary brain metastases.
Collapse
|
32
|
Zhang M, Ye F, Su M, Cui M, Chen H, Ma X. The Prognostic Role of Peritumoral Edema in Patients with Newly Diagnosed Glioblastoma: A Retrospective Analysis. J Clin Neurosci 2021; 89:249-257. [PMID: 34119276 DOI: 10.1016/j.jocn.2021.04.042] [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: 07/02/2020] [Revised: 04/29/2021] [Accepted: 04/30/2021] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Previous studies on glioblastomas (GBMs) have not reached a consensus on peritumoral edema (PTE)'s influence on survival. This study evaluated the PTE index's prognostic role in newly diagnosed GBMs using a well-designed method. METHODS Selected patients were reviewed after a rigorous screening process. Their general information was obtained from electronic medical records. The imaging metrics (MTD, TTM, TTE) representing tumor diameter, laterality, and PTE extent were obtained by manual measurement in Syngo FastView software. The PTE index was a ratio of TTE to MTD. Multiple variables were evaluated using analysis of variance and Cox regression model. RESULTS Of 143 patients, 62 were included in this study. MGMT promoter methylation and tumor laterality were both independent prognostic factors (p = 0.020, 0.042; HR = 0.272, 2.630). The lateral tumors' index was higher than that of the medial tumors (57.7% vs. 42.6%, p = 0.027). Low-index tumors were located in relatively medial positions compared with high-index tumors (TTM, 4.9 vs. 12.8, p = 0.032). This finding indicated that the PTE index tended to increase with tumor laterality. Moreover, the patients with low-index tumors had a significant survival disadvantage in the univariate analysis but not in the multivariate analysis (p = 0.023, 0.220). However, further analysis found that the combination of tumor laterality and PTE statistically stratified the survival outcome. The patients with lateral high-index tumors survived significantly longer (p = 0.022, HR = 1.927). CONCLUSIONS In contrast with the previous studies, this study recommends combining PTE and tumor laterality for survival stratification in newly diagnosed GBMs.
Collapse
Affiliation(s)
- Meng Zhang
- The Department of Neurosurgery, The First Medical Centre, Chinese PLA General Hospital, Fuxing Road 28, Haidian District, Beijing 100853, China; The Department of Neurosurgery, The Second Hospital of Southern District of Chinese Navy, Sanya Bay Road 82, Tianya District, Sanya 572000, China.
| | - Fuyue Ye
- The Department of Neurosurgery, The First Affiliated Hospital of Hainan Medical University, Longhua Road 31, Longhua District, Haikou 570102, China
| | - Meng Su
- The Department of Neurosurgery, The First Medical Centre, Chinese PLA General Hospital, Fuxing Road 28, Haidian District, Beijing 100853, China
| | - Meng Cui
- The Department of Neurosurgery, The First Medical Centre, Chinese PLA General Hospital, Fuxing Road 28, Haidian District, Beijing 100853, China
| | - Hongzun Chen
- The Department of Neurosurgery, The Second Hospital of Southern District of Chinese Navy, Sanya Bay Road 82, Tianya District, Sanya 572000, China
| | - Xiaodong Ma
- The Department of Neurosurgery, The First Medical Centre, Chinese PLA General Hospital, Fuxing Road 28, Haidian District, Beijing 100853, China.
| |
Collapse
|
33
|
Abstract
The 2016 World Health Organization brain tumor classification is based on genomic and molecular profile of tumor tissue. These characteristics have improved understanding of the brain tumor and played an important role in treatment planning and prognostication. There is an ongoing effort to develop noninvasive imaging techniques that provide insight into tissue characteristics at the cellular and molecular levels. This article focuses on the molecular characteristics of gliomas, transcriptomic subtypes, and radiogenomic studies using semantic and radiomic features. The limitations and future directions of radiogenomics as a standalone diagnostic tool also are discussed.
Collapse
Affiliation(s)
- Chaitra Badve
- Department of Radiology, Division of Neuroradiology, University Hospitals Cleveland Medical Center, BSH 5056, 11100 Euclid Avenue, Cleveland, OH 44106, USA.
| | - Sangam Kanekar
- Department of Radiology and Neurology, Division of Neuroradiology, Penn State College of Medicine, Penn State Milton Hershey Medical Center, Mail Code H066 500, University Drive, Hershey, PA 17033, USA
| |
Collapse
|
34
|
Estimating Glioblastoma Biophysical Growth Parameters Using Deep Learning Regression. BRAINLESION : GLIOMA, MULTIPLE SCLEROSIS, STROKE AND TRAUMATIC BRAIN INJURIES. BRAINLES (WORKSHOP) 2021; 12658:157-167. [PMID: 34514469 DOI: 10.1007/978-3-030-72084-1_15] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Glioblastoma ( GBM ) is arguably the most aggressive, infiltrative, and heterogeneous type of adult brain tumor. Biophysical modeling of GBM growth has contributed to more informed clinical decision-making. However, deploying a biophysical model to a clinical environment is challenging since underlying computations are quite expensive and can take several hours using existing technologies. Here we present a scheme to accelerate the computation. In particular, we present a deep learning ( DL )-based logistic regression model to estimate the GBM's biophysical growth in seconds. This growth is defined by three tumor-specific parameters: 1) a diffusion coefficient in white matter ( Dw ), which prescribes the rate of infiltration of tumor cells in white matter, 2) a mass-effect parameter ( Mp ), which defines the average tumor expansion, and 3) the estimated time ( T ) in number of days that the tumor has been growing. Preoperative structural multi-parametric MRI ( mpMRI ) scans from n = 135 subjects of the TCGA-GBM imaging collection are used to quantitatively evaluate our approach. We consider the mpMRI intensities within the region defined by the abnormal FLAIR signal envelope for training one DL model for each of the tumor-specific growth parameters. We train and validate the DL-based predictions against parameters derived from biophysical inversion models. The average Pearson correlation coefficients between our DL-based estimations and the biophysical parameters are 0.85 for Dw, 0.90 for Mp, and 0.94 for T, respectively. This study unlocks the power of tumor-specific parameters from biophysical tumor growth estimation. It paves the way towards their clinical translation and opens the door for leveraging advanced radiomic descriptors in future studies by means of a significantly faster parameter reconstruction compared to biophysical growth modeling approaches.
Collapse
|
35
|
Menze B, Isensee F, Wiest R, Wiestler B, Maier-Hein K, Reyes M, Bakas S. Analyzing magnetic resonance imaging data from glioma patients using deep learning. Comput Med Imaging Graph 2021; 88:101828. [PMID: 33571780 PMCID: PMC8040671 DOI: 10.1016/j.compmedimag.2020.101828] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 10/29/2020] [Accepted: 11/18/2020] [Indexed: 12/21/2022]
Abstract
The quantitative analysis of images acquired in the diagnosis and treatment of patients with brain tumors has seen a significant rise in the clinical use of computational tools. The underlying technology to the vast majority of these tools are machine learning methods and, in particular, deep learning algorithms. This review offers clinical background information of key diagnostic biomarkers in the diagnosis of glioma, the most common primary brain tumor. It offers an overview of publicly available resources and datasets for developing new computational tools and image biomarkers, with emphasis on those related to the Multimodal Brain Tumor Segmentation (BraTS) Challenge. We further offer an overview of the state-of-the-art methods in glioma image segmentation, again with an emphasis on publicly available tools and deep learning algorithms that emerged in the context of the BraTS challenge.
Collapse
Affiliation(s)
- Bjoern Menze
- Quantitative Biomedicine, University of Zurich, Zurich, Switzerland.
| | | | - Roland Wiest
- Support Center for Advanced Neuroimaging, Institute of Diagnostic and Interventional Neuroradiology, Inselspital, Bern, Switzerland.
| | | | | | | | - Spyridon Bakas
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
36
|
Venet L, Pati S, Feldman MD, Nasrallah MP, Yushkevich P, Bakas S. Accurate and Robust Alignment of Differently Stained Histologic Images Based on Greedy Diffeomorphic Registration. APPLIED SCIENCES (BASEL, SWITZERLAND) 2021; 11:1892. [PMID: 34290888 PMCID: PMC8291745 DOI: 10.3390/app11041892] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Histopathologic assessment routinely provides rich microscopic information about tissue structure and disease process. However, the sections used are very thin, and essentially capture only 2D representations of a certain tissue sample. Accurate and robust alignment of sequentially cut 2D slices should contribute to more comprehensive assessment accounting for surrounding 3D information. Towards this end, we here propose a two-step diffeomorphic registration approach that aligns differently stained histology slides to each other, starting with an initial affine step followed by estimating a deformation field. It was quantitatively evaluated on ample (n = 481) and diverse data from the automatic non-rigid histological image registration challenge, where it was awarded the second rank. The obtained results demonstrate the ability of the proposed approach to robustly (average robustness = 0.9898) and accurately (average relative target registration error = 0.2%) align differently stained histology slices of various anatomical sites while maintaining reasonable computational efficiency (<1 min per registration). The method was developed by adapting a general-purpose registration algorithm designed for 3D radiographic scans and achieved consistently accurate results for aligning high-resolution 2D histologic images. Accurate alignment of histologic images can contribute to a better understanding of the spatial arrangement and growth patterns of cells, vessels, matrix, nerves, and immune cell interactions.
Collapse
Affiliation(s)
- Ludovic Venet
- Center for Biomedical Image Computing & Analytics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sarthak Pati
- Center for Biomedical Image Computing & Analytics, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pathology & Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael D. Feldman
- Department of Pathology & Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - MacLean P. Nasrallah
- Department of Pathology & Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Paul Yushkevich
- Center for Biomedical Image Computing & Analytics, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Spyridon Bakas
- Center for Biomedical Image Computing & Analytics, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pathology & Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
37
|
Park CJ, Han K, Kim H, Ahn SS, Choi D, Park YW, Chang JH, Kim SH, Cha S, Lee SK. MRI Features May Predict Molecular Features of Glioblastoma in Isocitrate Dehydrogenase Wild-Type Lower-Grade Gliomas. AJNR Am J Neuroradiol 2021; 42:448-456. [PMID: 33509914 DOI: 10.3174/ajnr.a6983] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 10/19/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND PURPOSE Isocitrate dehydrogenase (IDH) wild-type lower-grade gliomas (histologic grades II and III) with epidermal growth factor receptor (EGFR) amplification or telomerase reverse transcriptase (TERT) promoter mutation are reported to behave similar to glioblastoma. We aimed to evaluate whether MR imaging features could identify a subset of IDH wild-type lower-grade gliomas that carry molecular features of glioblastoma. MATERIALS AND METHODS In this multi-institutional retrospective study, pathologically confirmed IDH wild-type lower-grade gliomas from 2 tertiary institutions and The Cancer Genome Atlas constituted the training set (institution 1 and The Cancer Genome Atlas, 64 patients) and the independent test set (institution 2, 57 patients). Preoperative MRIs were analyzed using the Visually AcceSAble Rembrandt Images and radiomics. The molecular glioblastoma status was determined on the basis of the presence of EGFR amplification and TERT promoter mutation. Molecular glioblastoma was present in 73.4% and 56.1% in the training and test sets, respectively. Models using clinical, Visually AcceSAble Rembrandt Images, and radiomic features were built to predict the molecular glioblastoma status in the training set; then they were validated in the test set. RESULTS In the test set, a model using both Visually AcceSAble Rembrandt Images and radiomic features showed superior predictive performance (area under the curve = 0.854) than that with only clinical features or Visually AcceSAble Rembrandt Images (areas under the curve = 0.514 and 0.648, respectively; P < . 001, both). When both Visually AcceSAble Rembrandt Images and radiomics were added to clinical features, the predictive performance significantly increased (areas under the curve = 0.514 versus 0.863, P < .001). CONCLUSIONS MR imaging features integrated with machine learning classifiers may predict a subset of IDH wild-type lower-grade gliomas that carry molecular features of glioblastoma.
Collapse
Affiliation(s)
- C J Park
- From the Department of Radiology (C.J.P.), Yonsei University College of Medicine, Seoul, Korea
| | - K Han
- Department of Radiology (K.H., H.K., S.S.A., Y.W.P., S.-K.L.), Research Institute of Radiological Sciences, Center for Clinical Imaging Data Science
| | - H Kim
- Department of Radiology (K.H., H.K., S.S.A., Y.W.P., S.-K.L.), Research Institute of Radiological Sciences, Center for Clinical Imaging Data Science
| | - S S Ahn
- Department of Radiology (K.H., H.K., S.S.A., Y.W.P., S.-K.L.), Research Institute of Radiological Sciences, Center for Clinical Imaging Data Science
| | - D Choi
- Department of Computer Science (D.C.), Yonsei University, Seoul, Korea
| | - Y W Park
- Department of Radiology (K.H., H.K., S.S.A., Y.W.P., S.-K.L.), Research Institute of Radiological Sciences, Center for Clinical Imaging Data Science
| | | | - S H Kim
- Department of Pathology (S.H.K.), Yonsei University College of Medicine, Seoul, Korea
| | - S Cha
- Department of Radiology and Biomedical Imaging (S.C.), University of California San Francisco, San Francisco, California
| | - S-K Lee
- Department of Radiology (K.H., H.K., S.S.A., Y.W.P., S.-K.L.), Research Institute of Radiological Sciences, Center for Clinical Imaging Data Science
| |
Collapse
|
38
|
Sanvito F, Castellano A, Falini A. Advancements in Neuroimaging to Unravel Biological and Molecular Features of Brain Tumors. Cancers (Basel) 2021; 13:cancers13030424. [PMID: 33498680 PMCID: PMC7865835 DOI: 10.3390/cancers13030424] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/15/2021] [Accepted: 01/19/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Advanced neuroimaging is gaining increasing relevance for the characterization and the molecular profiling of brain tumor tissue. On one hand, for some tumor types, the most widespread advanced techniques, investigating diffusion and perfusion features, have been proven clinically feasible and rather robust for diagnosis and prognosis stratification. In addition, 2-hydroxyglutarate spectroscopy, for the first time, offers the possibility to directly measure a crucial molecular marker. On the other hand, numerous innovative approaches have been explored for a refined evaluation of tumor microenvironments, particularly assessing microstructural and microvascular properties, and the potential applications of these techniques are vast and still to be fully explored. Abstract In recent years, the clinical assessment of primary brain tumors has been increasingly dependent on advanced magnetic resonance imaging (MRI) techniques in order to infer tumor pathophysiological characteristics, such as hemodynamics, metabolism, and microstructure. Quantitative radiomic data extracted from advanced MRI have risen as potential in vivo noninvasive biomarkers for predicting tumor grades and molecular subtypes, opening the era of “molecular imaging” and radiogenomics. This review presents the most relevant advancements in quantitative neuroimaging of advanced MRI techniques, by means of radiomics analysis, applied to primary brain tumors, including lower-grade glioma and glioblastoma, with a special focus on peculiar oncologic entities of current interest. Novel findings from diffusion MRI (dMRI), perfusion-weighted imaging (PWI), and MR spectroscopy (MRS) are hereby sifted in order to evaluate the role of quantitative imaging in neuro-oncology as a tool for predicting molecular profiles, stratifying prognosis, and characterizing tumor tissue microenvironments. Furthermore, innovative technological approaches are briefly addressed, including artificial intelligence contributions and ultra-high-field imaging new techniques. Lastly, after providing an overview of the advancements, we illustrate current clinical applications and future perspectives.
Collapse
Affiliation(s)
- Francesco Sanvito
- Neuroradiology Unit and CERMAC, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (F.S.); (A.F.)
- School of Medicine, Vita-Salute San Raffaele University, 20132 Milan, Italy
- Unit of Radiology, Department of Clinical, Surgical, Diagnostic, and Pediatric Sciences, University of Pavia, 27100 Pavia, Italy
| | - Antonella Castellano
- Neuroradiology Unit and CERMAC, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (F.S.); (A.F.)
- School of Medicine, Vita-Salute San Raffaele University, 20132 Milan, Italy
- Correspondence: ; Tel.: +39-02-2643-3015
| | - Andrea Falini
- Neuroradiology Unit and CERMAC, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (F.S.); (A.F.)
- School of Medicine, Vita-Salute San Raffaele University, 20132 Milan, Italy
| |
Collapse
|
39
|
Park JE, Kim HS, Lee J, Cheong EN, Shin I, Ahn SS, Shim WH. Deep-learned time-signal intensity pattern analysis using an autoencoder captures magnetic resonance perfusion heterogeneity for brain tumor differentiation. Sci Rep 2020; 10:21485. [PMID: 33293590 PMCID: PMC7723041 DOI: 10.1038/s41598-020-78485-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 11/11/2020] [Indexed: 01/10/2023] Open
Abstract
Current image processing methods for dynamic susceptibility contrast (DSC) magnetic resonance imaging (MRI) do not capture complex dynamic information of time-signal intensity curves. We investigated whether an autoencoder-based pattern analysis of DSC MRI captured representative temporal features that improves tissue characterization and tumor diagnosis in a multicenter setting. The autoencoder was applied to the time-signal intensity curves to obtain representative temporal patterns, which were subsequently learned by a convolutional neural network. This network was trained with 216 preoperative DSC MRI acquisitions and validated using external data (n = 43) collected with different DSC acquisition protocols. The autoencoder applied to time-signal intensity curves and clustering obtained nine representative clusters of temporal patterns, which accurately identified tumor and non-tumoral tissues. The dominant clusters of temporal patterns distinguished primary central nervous system lymphoma (PCNSL) from glioblastoma (AUC 0.89) and metastasis from glioblastoma (AUC 0.95). The autoencoder captured DSC time-signal intensity patterns that improved identification of tumoral tissues and differentiation of tumor type and was generalizable across centers.
Collapse
Affiliation(s)
- Ji Eun Park
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, 43 Olympic-ro 88, Songpa-Gu, Seoul, 05505, Korea
| | - Ho Sung Kim
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, 43 Olympic-ro 88, Songpa-Gu, Seoul, 05505, Korea.
| | - Junkyu Lee
- Department of Medical Science and Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, 43 Olympic-ro 88, Songpa-Gu, Seoul, Korea
| | - E-Nae Cheong
- Department of Medical Science and Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, 43 Olympic-ro 88, Songpa-Gu, Seoul, Korea
| | - Ilah Shin
- Department of Radiology, Research Institute of Radiological Science and Center for Clinical Imaging Data Science, Yonsei University College of Medicine, Seoul, Korea
| | - Sung Soo Ahn
- Department of Radiology, Research Institute of Radiological Science and Center for Clinical Imaging Data Science, Yonsei University College of Medicine, Seoul, Korea
| | - Woo Hyun Shim
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, 43 Olympic-ro 88, Songpa-Gu, Seoul, 05505, Korea.,Department of Medical Science and Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, 43 Olympic-ro 88, Songpa-Gu, Seoul, Korea
| |
Collapse
|
40
|
Pati S, Verma R, Akbari H, Bilello M, Hill VB, Sako C, Correa R, Beig N, Venet L, Thakur S, Serai P, Ha SM, Blake GD, Shinohara RT, Tiwari P, Bakas S. Reproducibility analysis of multi-institutional paired expert annotations and radiomic features of the Ivy Glioblastoma Atlas Project (Ivy GAP) dataset. Med Phys 2020; 47:6039-6052. [PMID: 33118182 DOI: 10.1002/mp.14556] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 07/26/2020] [Accepted: 08/26/2020] [Indexed: 12/15/2022] Open
Abstract
PURPOSE The availability of radiographic magnetic resonance imaging (MRI) scans for the Ivy Glioblastoma Atlas Project (Ivy GAP) has opened up opportunities for development of radiomic markers for prognostic/predictive applications in glioblastoma (GBM). In this work, we address two critical challenges with regard to developing robust radiomic approaches: (a) the lack of availability of reliable segmentation labels for glioblastoma tumor sub-compartments (i.e., enhancing tumor, non-enhancing tumor core, peritumoral edematous/infiltrated tissue) and (b) identifying "reproducible" radiomic features that are robust to segmentation variability across readers/sites. ACQUISITION AND VALIDATION METHODS From TCIA's Ivy GAP cohort, we obtained a paired set (n = 31) of expert annotations approved by two board-certified neuroradiologists at the Hospital of the University of Pennsylvania (UPenn) and at Case Western Reserve University (CWRU). For these studies, we performed a reproducibility study that assessed the variability in (a) segmentation labels and (b) radiomic features, between these paired annotations. The radiomic variability was assessed on a comprehensive panel of 11 700 radiomic features including intensity, volumetric, morphologic, histogram-based, and textural parameters, extracted for each of the paired sets of annotations. Our results demonstrated (a) a high level of inter-rater agreement (median value of DICE ≥0.8 for all sub-compartments), and (b) ≈24% of the extracted radiomic features being highly correlated (based on Spearman's rank correlation coefficient) to annotation variations. These robust features largely belonged to morphology (describing shape characteristics), intensity (capturing intensity profile statistics), and COLLAGE (capturing heterogeneity in gradient orientations) feature families. DATA FORMAT AND USAGE NOTES We make publicly available on TCIA's Analysis Results Directory (https://doi.org/10.7937/9j41-7d44), the complete set of (a) multi-institutional expert annotations for the tumor sub-compartments, (b) 11 700 radiomic features, and (c) the associated reproducibility meta-analysis. POTENTIAL APPLICATIONS The annotations and the associated meta-data for Ivy GAP are released with the purpose of enabling researchers toward developing image-based biomarkers for prognostic/predictive applications in GBM.
Collapse
Affiliation(s)
- Sarthak Pati
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, 19104, USA.,Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Ruchika Verma
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Hamed Akbari
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, 19104, USA.,Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Michel Bilello
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Virginia B Hill
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Chiharu Sako
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, 19104, USA.,Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Ramon Correa
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Niha Beig
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Ludovic Venet
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Siddhesh Thakur
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Prashant Serai
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, 19104, USA.,Department of Computer Science and Engineering, The Ohio State University, OH, 43210, USA
| | - Sung Min Ha
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Geri D Blake
- University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Russell Taki Shinohara
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, 19104, USA.,Penn Statistical Imaging and Visualization Endeavor (PennSIVE), University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Pallavi Tiwari
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Spyridon Bakas
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, 19104, USA.,Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.,Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
41
|
Beig N, Bera K, Tiwari P. Introduction to radiomics and radiogenomics in neuro-oncology: implications and challenges. Neurooncol Adv 2020; 2:iv3-iv14. [PMID: 33521636 PMCID: PMC7829475 DOI: 10.1093/noajnl/vdaa148] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Neuro-oncology largely consists of malignancies of the brain and central nervous system including both primary as well as metastatic tumors. Currently, a significant clinical challenge in neuro-oncology is to tailor therapies for patients based on a priori knowledge of their survival outcome or treatment response to conventional or experimental therapies. Radiomics or the quantitative extraction of subvisual data from conventional radiographic imaging has recently emerged as a powerful data-driven approach to offer insights into clinically relevant questions related to diagnosis, prediction, prognosis, as well as assessing treatment response. Furthermore, radiogenomic approaches provide a mechanism to establish statistical correlations of radiomic features with point mutations and next-generation sequencing data to further leverage the potential of routine MRI scans to serve as "virtual biopsy" maps. In this review, we provide an introduction to radiomic and radiogenomic approaches in neuro-oncology, including a brief description of the workflow involving preprocessing, tumor segmentation, and extraction of "hand-crafted" features from the segmented region of interest, as well as identifying radiogenomic associations that could ultimately lead to the development of reliable prognostic and predictive models in neuro-oncology applications. Lastly, we discuss the promise of radiomics and radiogenomic approaches in personalizing treatment decisions in neuro-oncology, as well as the challenges with clinical adoption, which will rely heavily on their demonstrated resilience to nonstandardization in imaging protocols across sites and scanners, as well as in their ability to demonstrate reproducibility across large multi-institutional cohorts.
Collapse
Affiliation(s)
- Niha Beig
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Kaustav Bera
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Pallavi Tiwari
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
42
|
Hu J, Xiao Q, Dong M, Guo D, Wu X, Wang B. Glioblastoma Immunotherapy Targeting the Innate Immune Checkpoint CD47-SIRPα Axis. Front Immunol 2020; 11:593219. [PMID: 33329583 PMCID: PMC7728717 DOI: 10.3389/fimmu.2020.593219] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 11/02/2020] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma Multiforme (GBM) is the most common and aggressive form of intracranial tumors with poor prognosis. In recent years, tumor immunotherapy has been an attractive strategy for a variety of tumors. Currently, most immunotherapies take advantage of the adaptive anti-tumor immunity, such as cytotoxic T cells. However, the predominant accumulation of tumor-associated microglia/macrophages (TAMs) results in limited success of these strategies in the glioblastoma. To improve the immunotherapeutic efficacy for GBM, it is detrimental to understand the role of TAM in glioblastoma immunosuppressive microenvironment. In this review, we will discuss the roles of CD47-SIRPα axis in TAMs infiltration and activities and the promising effects of targeting this axis on the activation of both innate and adaptive antitumor immunity in glioblastoma.
Collapse
Affiliation(s)
- Jinyang Hu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qungen Xiao
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Minhai Dong
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dongsheng Guo
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xudong Wu
- Department of Cell Biology, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Key Laboratory of Medical Epigenetics, Tianjin Medical University, Tianjin, China.,Department of Neurosurgery, Tianjin Medical University General Hospital and Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, China
| | - Baofeng Wang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
43
|
Farrell C, Shi W, Bodman A, Olson JJ. Congress of neurological surgeons systematic review and evidence-based guidelines update on the role of emerging developments in the management of newly diagnosed glioblastoma. J Neurooncol 2020; 150:269-359. [PMID: 33215345 DOI: 10.1007/s11060-020-03607-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 08/23/2020] [Indexed: 12/12/2022]
Abstract
TARGET POPULATION These recommendations apply to adult patients with newly diagnosed or suspected glioblastoma. IMAGING Question What imaging modalities are in development that may be able to provide improvements in diagnosis, and therapeutic guidance for individuals with newly diagnosed glioblastoma? RECOMMENDATION Level III: It is suggested that techniques utilizing magnetic resonance imaging for diffusion weighted imaging, and to measure cerebral blood and magnetic spectroscopic resonance imaging of N-acetyl aspartate, choline and the choline to N-acetyl aspartate index to assist in diagnosis and treatment planning in patients with newly diagnosed or suspected glioblastoma. SURGERY Question What new surgical techniques can be used to provide improved tumor definition and resectability to yield better tumor control and prognosis for individuals with newly diagnosed glioblastoma? RECOMMENDATIONS Level II: The use of 5-aminolevulinic acid is recommended to improve extent of tumor resection in patients with newly diagnosed glioblastoma. Level II: The use of 5-aminolevulinic acid is recommended to improve median survival and 2 year survival in newly diagnosed glioblastoma patients with clinical characteristics suggesting poor prognosis. Level III: It is suggested that, when available, patients be enrolled in properly designed clinical trials assessing the value of diffusion tensor imaging in improving the safety of patients with newly diagnosed glioblastoma undergoing surgery. NEUROPATHOLOGY Question What new pathology techniques and measurement of biomarkers in tumor tissue can be used to provide improved diagnostic ability, and determination of therapeutic responsiveness and prognosis for patients with newly diagnosed glioblastomas? RECOMMENDATIONS Level II: Assessment of tumor MGMT promoter methylation status is recommended as a significant predictor of a longer progression free survival and overall survival in patients with newly diagnosed with glioblastoma. Level II: Measurement of tumor expression of neuron-glia-2, neurofilament protein, glutamine synthetase and phosphorylated STAT3 is recommended as a predictor of overall survival in patients with newly diagnosed with glioblastoma. Level III: Assessment of tumor IDH1 mutation status is suggested as a predictor of longer progression free survival and overall survival in patients with newly diagnosed with glioblastoma. Level III: Evaluation of tumor expression of Phosphorylated Mitogen-Activated Protein Kinase protein, EGFR protein, and Insulin-like Growth Factor-Binding Protein-3 is suggested as a predictor of overall survival in patients with newly diagnosed with glioblastoma. RADIATION Question What radiation therapy techniques are in development that may be used to provide improved tumor control and prognosis for individuals with newly diagnosed glioblastomas? RECOMMENDATIONS Level III: It is suggested that patients with newly diagnosed glioblastoma undergo pretreatment radio-labeled amino acid tracer positron emission tomography to assess areas at risk for tumor recurrence to assist in radiation treatment planning. Level III: It is suggested that, when available, patients be with newly diagnosed glioblastomas be enrolled in properly designed clinical trials of radiation dose escalation, altered fractionation, or new radiation delivery techniques. CHEMOTHERAPY Question What emerging chemotherapeutic agents or techniques are available to provide better tumor control and prognosis for patients with newly diagnosed glioblastomas? RECOMMENDATION Level III: As no emerging chemotherapeutic agents or techniques were identified in this review that improved tumor control and prognosis it is suggested that, when available, patients with newly diagnosed glioblastomas be enrolled in properly designed clinical trials of chemotherapy. MOLECULAR AND TARGETED THERAPY Question What new targeted therapy agents are available to provide better tumor control and prognosis for individuals with newly diagnosed glioblastomas? RECOMMENDATION Level III: As no new molecular and targeted therapies have clearly provided better tumor control and prognosis it is suggested that, when available, patients with newly diagnosed glioblastomas be enrolled in properly designed clinical trials of molecular and targeted therapies IMMUNOTHERAPY: Question What emerging immunotherapeutic agents or techniques are available to provide better tumor control and prognosis for patients with newly diagnosed glioblastomas? RECOMMENDATION Level III: As no immunotherapeutic agents have clearly provided better tumor control and prognosis it is suggested that, when available, patients with newly diagnosed glioblastomas be enrolled in properly designed clinical trials of immunologically-based therapies. NOVEL THERAPIES Question What novel therapies or techniques are in development to provide better tumor control and prognosis for individuals with newly diagnosed glioblastomas? RECOMMENDATIONS Level II: The use of tumor-treating fields is recommended for patients with newly diagnosed glioblastoma who have undergone surgical debulking and completed concurrent chemoradiation without progression of disease at the time of tumor-treating field therapy initiation. Level II: It is suggested that, when available, enrollment in properly designed studies of vector containing herpes simplex thymidine kinase gene and prodrug therapies be considered in patients with newly diagnosed glioblastoma.
Collapse
Affiliation(s)
- Christopher Farrell
- Department of Neurosurgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Wenyin Shi
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA, USA
| | | | - Jeffrey J Olson
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
44
|
Fathi Kazerooni A, Akbari H, Shukla G, Badve C, Rudie JD, Sako C, Rathore S, Bakas S, Pati S, Singh A, Bergman M, Ha SM, Kontos D, Nasrallah M, Bagley SJ, Lustig RA, O'Rourke DM, Sloan AE, Barnholtz-Sloan JS, Mohan S, Bilello M, Davatzikos C. Cancer Imaging Phenomics via CaPTk: Multi-Institutional Prediction of Progression-Free Survival and Pattern of Recurrence in Glioblastoma. JCO Clin Cancer Inform 2020; 4:234-244. [PMID: 32191542 PMCID: PMC7113126 DOI: 10.1200/cci.19.00121] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
PURPOSE To construct a multi-institutional radiomic model that supports upfront prediction of progression-free survival (PFS) and recurrence pattern (RP) in patients diagnosed with glioblastoma multiforme (GBM) at the time of initial diagnosis. PATIENTS AND METHODS We retrospectively identified data for patients with newly diagnosed GBM from two institutions (institution 1, n = 65; institution 2, n = 15) who underwent gross total resection followed by standard adjuvant chemoradiation therapy, with pathologically confirmed recurrence, sufficient follow-up magnetic resonance imaging (MRI) scans to reliably determine PFS, and available presurgical multiparametric MRI (MP-MRI). The advanced software suite Cancer Imaging Phenomics Toolkit (CaPTk) was leveraged to analyze standard clinical brain MP-MRI scans. A rich set of imaging features was extracted from the MP-MRI scans acquired before the initial resection and was integrated into two distinct imaging signatures for predicting mean shorter or longer PFS and near or distant RP. The predictive signatures for PFS and RP were evaluated on the basis of different classification schemes: single-institutional analysis, multi-institutional analysis with random partitioning of the data into discovery and replication cohorts, and multi-institutional assessment with data from institution 1 as the discovery cohort and data from institution 2 as the replication cohort. RESULTS These predictors achieved cross-validated classification performance (ie, area under the receiver operating characteristic curve) of 0.88 (single-institution analysis) and 0.82 to 0.83 (multi-institution analysis) for prediction of PFS and 0.88 (single-institution analysis) and 0.56 to 0.71 (multi-institution analysis) for prediction of RP. CONCLUSION Imaging signatures of presurgical MP-MRI scans reveal relatively high predictability of time and location of GBM recurrence, subject to the patients receiving standard first-line chemoradiation therapy. Through its graphical user interface, CaPTk offers easy accessibility to advanced computational algorithms for deriving imaging signatures predictive of clinical outcome and could similarly be used for a variety of radiomic and radiogenomic analyses.
Collapse
Affiliation(s)
- Anahita Fathi Kazerooni
- Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.,Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Hamed Akbari
- Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.,Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Gaurav Shukla
- Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.,Department of Radiation Oncology, Christiana Care Helen F. Graham Cancer Center and Research Institute, Newark, DE.,Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Chaitra Badve
- Department of Radiology, University Hospitals-Seidman Cancer Center, Cleveland, OH.,Case Comprehensive Cancer Center, Cleveland, OH
| | - Jeffrey D Rudie
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.,Department of Radiology and Biomedical Imaging, University of California at San Francisco, San Francisco, CA
| | - Chiharu Sako
- Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.,Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Saima Rathore
- Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.,Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Spyridon Bakas
- Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.,Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.,Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Sarthak Pati
- Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.,Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Ashish Singh
- Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.,Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Mark Bergman
- Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.,Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Sung Min Ha
- Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.,Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Despina Kontos
- Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.,Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - MacLean Nasrallah
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Stephen J Bagley
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Robert A Lustig
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Donald M O'Rourke
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.,Glioblastoma Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Andrew E Sloan
- Case Western Reserve University School of Medicine, Cleveland, OH.,Case Comprehensive Cancer Center, Cleveland, OH.,Department of Neurologic Surgery, University Hospitals-Seidman Cancer Center, Cleveland, OH
| | - Jill S Barnholtz-Sloan
- Case Western Reserve University School of Medicine, Cleveland, OH.,Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, OH
| | - Suyash Mohan
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Michel Bilello
- Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.,Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Christos Davatzikos
- Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.,Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
45
|
Abstract
Magnetic resonance imaging (MRI) is a noninvasive imaging tool for neuroradiological diagnosis. Numerous concepts of automated MRI analysis and the use of machine learning have been proposed to assist diagnosis and prognosis. While these academic innovations have proven effective in principle within controlled environments, their application to clinical practice has faced unmet requirements, such as the ability to perform reliably across a heterogeneous population, to work robustly in the presence of comorbidities, and to be invariant to scanner hardware and image quality. The lack of realistic confidence bounds and the inability to handle missing data have also reduced the application of most of these methods outside of academic studies. Mastering the complex challenges in the diagnostic process may help researchers discover novel biological constructs in multimodal data and improve stratification for clinical trials, paving the way for precision medicine. This review presents the state of the art of computerized brain MRI analysis for diagnostic purposes. We critically evaluate the current clinical usefulness of the methods and highlight challenges and future perspectives of the field.
Collapse
Affiliation(s)
- Saima Rathore
- Center for Biomedical Image Computing and Analytics, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Ahmed Abdulkadir
- Center for Biomedical Image Computing and Analytics, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- University Hospital of of Old Age Psychiatry and Psychotherapy, University of Bern, 3008 Bern, Switzerland
| | - Christos Davatzikos
- Center for Biomedical Image Computing and Analytics, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
46
|
Bakas S, Shukla G, Akbari H, Erus G, Sotiras A, Rathore S, Sako C, Min Ha S, Rozycki M, Shinohara RT, Bilello M, Davatzikos C. Overall survival prediction in glioblastoma patients using structural magnetic resonance imaging (MRI): advanced radiomic features may compensate for lack of advanced MRI modalities. J Med Imaging (Bellingham) 2020; 7:031505. [PMID: 32566694 DOI: 10.1117/1.jmi.7.3.031505] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 05/20/2020] [Indexed: 12/22/2022] Open
Abstract
Purpose: Glioblastoma, the most common and aggressive adult brain tumor, is considered noncurative at diagnosis, with 14 to 16 months median survival following treatment. There is increasing evidence that noninvasive integrative analysis of radiomic features can predict overall and progression-free survival, using advanced multiparametric magnetic resonance imaging (Adv-mpMRI). If successfully applicable, such noninvasive markers can considerably influence patient management. However, most patients prior to initiation of therapy typically undergo only basic structural mpMRI (Bas-mpMRI, i.e., T1, T1-Gd, T2, and T2-fluid-attenuated inversion recovery) preoperatively, rather than Adv-mpMRI that provides additional vascularization (dynamic susceptibility contrast-MRI) and cell-density (diffusion tensor imaging) related information. Approach: We assess a retrospective cohort of 101 glioblastoma patients with available Adv-mpMRI from a previous study, which has shown that an initial feature panel (IFP, i.e., intensity, volume, location, and growth model parameters) extracted from Adv-mpMRI can yield accurate overall survival stratification. We focus on demonstrating that equally accurate prediction models can be constructed using augmented radiomic feature panels (ARFPs, i.e., integrating morphology and textural descriptors) extracted solely from widely available Bas-mpMRI, obviating the need for using Adv-mpMRI. We extracted 1612 radiomic features from distinct tumor subregions to build multivariate models that stratified patients as long-, intermediate-, or short-survivors. Results: The classification accuracy of the model utilizing Adv-mpMRI protocols and the IFP was 72.77% and degraded to 60.89% when using only Bas-mpMRI. However, utilizing the ARFP on Bas-mpMRI improved the accuracy to 74.26%. Furthermore, Kaplan-Meier analysis demonstrated superior classification of subjects into short-, intermediate-, and long-survivor classes when using ARFP extracted from Bas-mpMRI. Conclusions: This quantitative evaluation indicates that accurate survival prediction in glioblastoma patients is feasible using solely Bas-mpMRI and integrative advanced radiomic features, which can compensate for the lack of Adv-mpMRI. Our finding holds promise for generalization across multiple institutions that may not have access to Adv-mpMRI and to better inform clinical decision-making about aggressive interventions and clinical trials.
Collapse
Affiliation(s)
- Spyridon Bakas
- University of Pennsylvania, Perelman School of Medicine, Center for Biomedical Image Computing and Analytics, Richards Medical Research Laboratories, Philadelphia, PA, United States.,University of Pennsylvania, Perelman School of Medicine, Richards Medical Research Laboratories, Department of Radiology, Philadelphia, PA, United States.,University of Pennsylvania, Perelman School of Medicine, Richards Medical Research Laboratories, Department of Pathology and Laboratory Medicine, Philadelphia, PA, United States
| | - Gaurav Shukla
- University of Pennsylvania, Perelman School of Medicine, Center for Biomedical Image Computing and Analytics, Richards Medical Research Laboratories, Philadelphia, PA, United States.,Thomas Jefferson University, Sidney Kimmel Cancer Center, Department of Radiation Oncology, Philadelphia, PA, United States
| | - Hamed Akbari
- University of Pennsylvania, Perelman School of Medicine, Center for Biomedical Image Computing and Analytics, Richards Medical Research Laboratories, Philadelphia, PA, United States.,University of Pennsylvania, Perelman School of Medicine, Richards Medical Research Laboratories, Department of Radiology, Philadelphia, PA, United States
| | - Guray Erus
- University of Pennsylvania, Perelman School of Medicine, Center for Biomedical Image Computing and Analytics, Richards Medical Research Laboratories, Philadelphia, PA, United States.,University of Pennsylvania, Perelman School of Medicine, Richards Medical Research Laboratories, Department of Radiology, Philadelphia, PA, United States
| | - Aristeidis Sotiras
- University of Pennsylvania, Perelman School of Medicine, Center for Biomedical Image Computing and Analytics, Richards Medical Research Laboratories, Philadelphia, PA, United States.,University of Pennsylvania, Perelman School of Medicine, Richards Medical Research Laboratories, Department of Radiology, Philadelphia, PA, United States.,Washington University in St. Louis, School of Medicine, Institute for Informatics, Saint Louis, MO, United States.,Washington University in St. Louis, Department of Radiology, Saint Louis, MO, United States
| | - Saima Rathore
- University of Pennsylvania, Perelman School of Medicine, Center for Biomedical Image Computing and Analytics, Richards Medical Research Laboratories, Philadelphia, PA, United States.,University of Pennsylvania, Perelman School of Medicine, Richards Medical Research Laboratories, Department of Radiology, Philadelphia, PA, United States
| | - Chiharu Sako
- University of Pennsylvania, Perelman School of Medicine, Center for Biomedical Image Computing and Analytics, Richards Medical Research Laboratories, Philadelphia, PA, United States.,University of Pennsylvania, Perelman School of Medicine, Richards Medical Research Laboratories, Department of Radiology, Philadelphia, PA, United States
| | - Sung Min Ha
- University of Pennsylvania, Perelman School of Medicine, Center for Biomedical Image Computing and Analytics, Richards Medical Research Laboratories, Philadelphia, PA, United States.,University of Pennsylvania, Perelman School of Medicine, Richards Medical Research Laboratories, Department of Radiology, Philadelphia, PA, United States
| | - Martin Rozycki
- University of Pennsylvania, Perelman School of Medicine, Center for Biomedical Image Computing and Analytics, Richards Medical Research Laboratories, Philadelphia, PA, United States.,University of Pennsylvania, Perelman School of Medicine, Richards Medical Research Laboratories, Department of Radiology, Philadelphia, PA, United States
| | - Russell T Shinohara
- University of Pennsylvania, Perelman School of Medicine, Center for Biomedical Image Computing and Analytics, Richards Medical Research Laboratories, Philadelphia, PA, United States.,University of Pennsylvania, Perelman School of Medicine, Penn Statistics in Imaging and Visualization Center, Department of Biostatistics, Epidemiology, and Informatics, Philadelphia, PA, United States
| | - Michel Bilello
- University of Pennsylvania, Perelman School of Medicine, Center for Biomedical Image Computing and Analytics, Richards Medical Research Laboratories, Philadelphia, PA, United States.,University of Pennsylvania, Perelman School of Medicine, Richards Medical Research Laboratories, Department of Radiology, Philadelphia, PA, United States
| | - Christos Davatzikos
- University of Pennsylvania, Perelman School of Medicine, Center for Biomedical Image Computing and Analytics, Richards Medical Research Laboratories, Philadelphia, PA, United States.,University of Pennsylvania, Perelman School of Medicine, Richards Medical Research Laboratories, Department of Radiology, Philadelphia, PA, United States
| |
Collapse
|
47
|
Mang A, Bakas S, Subramanian S, Davatzikos C, Biros G. Integrated Biophysical Modeling and Image Analysis: Application to Neuro-Oncology. Annu Rev Biomed Eng 2020; 22:309-341. [PMID: 32501772 PMCID: PMC7520881 DOI: 10.1146/annurev-bioeng-062117-121105] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Central nervous system (CNS) tumors come with vastly heterogeneous histologic, molecular, and radiographic landscapes, rendering their precise characterization challenging. The rapidly growing fields of biophysical modeling and radiomics have shown promise in better characterizing the molecular, spatial, and temporal heterogeneity of tumors. Integrative analysis of CNS tumors, including clinically acquired multi-parametric magnetic resonance imaging (mpMRI) and the inverse problem of calibrating biophysical models to mpMRI data, assists in identifying macroscopic quantifiable tumor patterns of invasion and proliferation, potentially leading to improved (a) detection/segmentation of tumor subregions and (b) computer-aided diagnostic/prognostic/predictive modeling. This article presents a summary of (a) biophysical growth modeling and simulation,(b) inverse problems for model calibration, (c) these models' integration with imaging workflows, and (d) their application to clinically relevant studies. We anticipate that such quantitative integrative analysis may even be beneficial in a future revision of the World Health Organization (WHO) classification for CNS tumors, ultimately improving patient survival prospects.
Collapse
Affiliation(s)
- Andreas Mang
- Department of Mathematics, University of Houston, Houston, Texas 77204, USA;
| | - Spyridon Bakas
- Department of Mathematics, University of Houston, Houston, Texas 77204, USA;
| | - Shashank Subramanian
- Oden Institute of Computational Engineering and Sciences, The University of Texas at Austin, Austin, Texas 78712, USA; ,
| | - Christos Davatzikos
- Center for Biomedical Image Computing and Analytics (CBICA); Department of Radiology; and Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA; ,
| | - George Biros
- Oden Institute of Computational Engineering and Sciences, The University of Texas at Austin, Austin, Texas 78712, USA; ,
| |
Collapse
|
48
|
Grenko CM, Viaene AN, Nasrallah MP, Feldman MD, Akbari H, Bakas S. Towards Population-Based Histologic Stain Normalization of Glioblastoma. ACTA ACUST UNITED AC 2020; 11992:44-56. [PMID: 32743562 DOI: 10.1007/978-3-030-46640-4_5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Glioblastoma ( 'GBM' ) is the most aggressive type of primary malignant adult brain tumor, with very heterogeneous radio-graphic, histologic, and molecular profiles. A growing body of advanced computational analyses are conducted towards further understanding the biology and variation in glioblastoma. To address the intrinsic heterogeneity among different computational studies, reference standards have been established to facilitate both radiographic and molecular analyses, e.g., anatomical atlas for image registration and housekeeping genes, respectively. However, there is an apparent lack of reference standards in the domain of digital pathology, where each independent study uses an arbitrarily chosen slide from their evaluation dataset for normalization purposes. In this study, we introduce a novel stain normalization approach based on a composite reference slide comprised of information from a large population of anatomically annotated hematoxylin and eosin ( 'H&E' ) whole-slide images from the Ivy Glioblastoma Atlas Project ( 'IvyGAP' ). Two board-certified neuropathologists manually reviewed and selected annotations in 509 slides, according to the World Health Organization definitions. We computed summary statistics from each of these approved annotations and weighted them based on their percent contribution to overall slide ( 'PCOS' ), to form a global histogram and stain vectors. Quantitative evaluation of pre- and post-normalization stain density statistics for each annotated region with PCOS > 0.05% yielded a significant (largest p = 0.001, two-sided Wilcoxon rank sum test) reduction of its intensity variation for both 'H' & 'E' . Subject to further large-scale evaluation, our findings support the proposed approach as a potentially robust population-based reference for stain normalization.
Collapse
Affiliation(s)
- Caleb M Grenko
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA.,Center for Interdisciplinary Studies, Davidson College, Davidson, NC, USA
| | - Angela N Viaene
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, USA
| | - MacLean P Nasrallah
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael D Feldman
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hamed Akbari
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA.,Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Spyridon Bakas
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA.,Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
49
|
Pati S, Singh A, Rathore S, Gastounioti A, Bergman M, Ngo P, Ha SM, Bounias D, Minock J, Murphy G, Li H, Bhattarai A, Wolf A, Sridaran P, Kalarot R, Akbari H, Sotiras A, Thakur SP, Verma R, Shinohara RT, Yushkevich P, Fan Y, Kontos D, Davatzikos C, Bakas S. The Cancer Imaging Phenomics Toolkit (CaPTk): Technical Overview. BRAINLESION : GLIOMA, MULTIPLE SCLEROSIS, STROKE AND TRAUMATIC BRAIN INJURIES. BRAINLES (WORKSHOP) 2020; 11993:380-394. [PMID: 32754723 PMCID: PMC7402244 DOI: 10.1007/978-3-030-46643-5_38] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The purpose of this manuscript is to provide an overview of the technical specifications and architecture of the Cancer imaging Phenomics Toolkit (CaPTk www.cbica.upenn.edu/captk), a cross-platform, open-source, easy-to-use, and extensible software platform for analyzing 2D and 3D images, currently focusing on radiographic scans of brain, breast, and lung cancer. The primary aim of this platform is to enable swift and efficient translation of cutting-edge academic research into clinically useful tools relating to clinical quantification, analysis, predictive modeling, decision-making, and reporting workflow. CaPTk builds upon established open-source software toolkits, such as the Insight Toolkit (ITK) and OpenCV, to bring together advanced computational functionality. This functionality describes specialized, as well as general-purpose, image analysis algorithms developed during active multi-disciplinary collaborative research studies to address real clinical requirements. The target audience of CaPTk consists of both computational scientists and clinical experts. For the former it provides i) an efficient image viewer offering the ability of integrating new algorithms, and ii) a library of readily-available clinically-relevant algorithms, allowing batch-processing of multiple subjects. For the latter it facilitates the use of complex algorithms for clinically-relevant studies through a user-friendly interface, eliminating the prerequisite of a substantial computational background. CaPTk's long-term goal is to provide widely-used technology to make use of advanced quantitative imaging analytics in cancer prediction, diagnosis and prognosis, leading toward a better understanding of the biological mechanisms of cancer development.
Collapse
Affiliation(s)
- Sarthak Pati
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
| | - Ashish Singh
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
| | - Saima Rathore
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Aimilia Gastounioti
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mark Bergman
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
| | - Phuc Ngo
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sung Min Ha
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Dimitrios Bounias
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
| | - James Minock
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
| | - Grayson Murphy
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
| | - Hongming Li
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amit Bhattarai
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
| | - Adam Wolf
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
| | - Patmaa Sridaran
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
| | - Ratheesh Kalarot
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
| | - Hamed Akbari
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Aristeidis Sotiras
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology and Institute for Informatics, School of Medicine, Washington University in St. Louis, Saint Louis, MO, USA
| | - Siddhesh P Thakur
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
| | - Ragini Verma
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Russell T Shinohara
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Penn Statistics in Imaging and Visualization Endeavor (PennSIVE), University of Pennsylvania, Philadelphia, PA, USA
| | - Paul Yushkevich
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn Image Computing and Science Lab., University of Pennsylvania (PICSL), Philadelphia, PA, USA
| | - Yong Fan
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Despina Kontos
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christos Davatzikos
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Spyridon Bakas
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
50
|
Beig N, Bera K, Prasanna P, Antunes J, Correa R, Singh S, Saeed Bamashmos A, Ismail M, Braman N, Verma R, Hill VB, Statsevych V, Ahluwalia MS, Varadan V, Madabhushi A, Tiwari P. Radiogenomic-Based Survival Risk Stratification of Tumor Habitat on Gd-T1w MRI Is Associated with Biological Processes in Glioblastoma. Clin Cancer Res 2020; 26:1866-1876. [PMID: 32079590 PMCID: PMC7165059 DOI: 10.1158/1078-0432.ccr-19-2556] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 10/11/2019] [Accepted: 01/14/2020] [Indexed: 01/31/2023]
Abstract
PURPOSE To (i) create a survival risk score using radiomic features from the tumor habitat on routine MRI to predict progression-free survival (PFS) in glioblastoma and (ii) obtain a biological basis for these prognostic radiomic features, by studying their radiogenomic associations with molecular signaling pathways. EXPERIMENTAL DESIGN Two hundred three patients with pretreatment Gd-T1w, T2w, T2w-FLAIR MRI were obtained from 3 cohorts: The Cancer Imaging Archive (TCIA; n = 130), Ivy GAP (n = 32), and Cleveland Clinic (n = 41). Gene-expression profiles of corresponding patients were obtained for TCIA cohort. For every study, following expert segmentation of tumor subcompartments (necrotic core, enhancing tumor, peritumoral edema), 936 3D radiomic features were extracted from each subcompartment across all MRI protocols. Using Cox regression model, radiomic risk score (RRS) was developed for every protocol to predict PFS on the training cohort (n = 130) and evaluated on the holdout cohort (n = 73). Further, Gene Ontology and single-sample gene set enrichment analysis were used to identify specific molecular signaling pathway networks associated with RRS features. RESULTS Twenty-five radiomic features from the tumor habitat yielded the RRS. A combination of RRS with clinical (age and gender) and molecular features (MGMT and IDH status) resulted in a concordance index of 0.81 (P < 0.0001) on training and 0.84 (P = 0.03) on the test set. Radiogenomic analysis revealed associations of RRS features with signaling pathways for cell differentiation, cell adhesion, and angiogenesis, which contribute to chemoresistance in GBM. CONCLUSIONS Our findings suggest that prognostic radiomic features from routine Gd-T1w MRI may also be significantly associated with key biological processes that affect response to chemotherapy in GBM.
Collapse
Affiliation(s)
- Niha Beig
- Case Western Reserve University, Cleveland, Ohio
| | - Kaustav Bera
- Case Western Reserve University, Cleveland, Ohio
| | | | | | - Ramon Correa
- Case Western Reserve University, Cleveland, Ohio
| | | | | | - Marwa Ismail
- Case Western Reserve University, Cleveland, Ohio
| | | | | | - Virginia B Hill
- Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | | | | | | | - Anant Madabhushi
- Case Western Reserve University, Cleveland, Ohio
- Louis Stokes Cleveland Veterans Administration Medical Center, Cleveland, Ohio
| | | |
Collapse
|