1
|
Akbari H, Bakas S, Sako C, Fathi Kazerooni A, Villanueva-Meyer J, Garcia JA, Mamourian E, Liu F, Cao Q, Shinohara RT, Baid U, Getka A, Pati S, Singh A, Calabrese E, Chang S, Rudie J, Sotiras A, LaMontagne P, Marcus DS, Milchenko M, Nazeri A, Balana C, Capellades J, Puig J, Badve C, Barnholtz-Sloan JS, Sloan AE, Vadmal V, Waite K, Ak M, Colen RR, Park YW, Ahn SS, Chang JH, Choi YS, Lee SK, Alexander GS, Ali AS, Dicker AP, Flanders AE, Liem S, Lombardo J, Shi W, Shukla G, Griffith B, Poisson LM, Rogers LR, Kotrotsou A, Booth TC, Jain R, Lee M, Mahajan A, Chakravarti A, Palmer JD, DiCostanzo D, Fathallah-Shaykh H, Cepeda S, Santonocito OS, Di Stefano AL, Wiestler B, Melhem ER, Woodworth GF, Tiwari P, Valdes P, Matsumoto Y, Otani Y, Imoto R, Aboian M, Koizumi S, Kurozumi K, Kawakatsu T, Alexander K, Satgunaseelan L, Rulseh AM, Bagley SJ, Bilello M, Binder ZA, Brem S, Desai AS, Lustig RA, Maloney E, Prior T, Amankulor N, Nasrallah MP, O’Rourke DM, Mohan S, Davatzikos C. Machine learning-based prognostic subgrouping of glioblastoma: A multicenter study. Neuro Oncol 2025; 27:1102-1115. [PMID: 39665363 PMCID: PMC12083074 DOI: 10.1093/neuonc/noae260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Indexed: 12/13/2024] Open
Abstract
BACKGROUND Glioblastoma (GBM) is the most aggressive adult primary brain cancer, characterized by significant heterogeneity, posing challenges for patient management, treatment planning, and clinical trial stratification. METHODS We developed a highly reproducible, personalized prognostication, and clinical subgrouping system using machine learning (ML) on routine clinical data, magnetic resonance imaging (MRI), and molecular measures from 2838 demographically diverse patients across 22 institutions and 3 continents. Patients were stratified into favorable, intermediate, and poor prognostic subgroups (I, II, and III) using Kaplan-Meier analysis (Cox proportional model and hazard ratios [HR]). RESULTS The ML model stratified patients into distinct prognostic subgroups with HRs between subgroups I-II and I-III of 1.62 (95% CI: 1.43-1.84, P < .001) and 3.48 (95% CI: 2.94-4.11, P < .001), respectively. Analysis of imaging features revealed several tumor properties contributing unique prognostic value, supporting the feasibility of a generalizable prognostic classification system in a diverse cohort. CONCLUSIONS Our ML model demonstrates extensive reproducibility and online accessibility, utilizing routine imaging data rather than complex imaging protocols. This platform offers a unique approach to personalized patient management and clinical trial stratification in GBM.
Collapse
Affiliation(s)
- Hamed Akbari
- Department of Bioengineering, School of Engineering, Santa Clara University, Santa Clara, California, USA
| | - Spyridon Bakas
- Department of Pathology & Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Computer Science, Luddy School of Informatics, Computing, and Engineering, Indiana University, Indianapolis, Indiana, USA
| | - Chiharu Sako
- Center for Data Science and AI for Integrated Diagnostics (AI2D), and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Penn Statistics in Imaging and Visualization Center, and Center for Clinical Epidemiology ce and AI for Integrated Diagnostics (AI2D), and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Anahita Fathi Kazerooni
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Center for Data-Driven Discovery in Biomedicine (D3b), Division of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Javier Villanueva-Meyer
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California, USA
| | - Jose A Garcia
- Center for Data Science and AI for Integrated Diagnostics (AI2D), and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Penn Statistics in Imaging and Visualization Center, and Center for Clinical Epidemiology ce and AI for Integrated Diagnostics (AI2D), and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Elizabeth Mamourian
- Center for Data Science and AI for Integrated Diagnostics (AI2D), and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Penn Statistics in Imaging and Visualization Center, and Center for Clinical Epidemiology ce and AI for Integrated Diagnostics (AI2D), and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Fang Liu
- Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Quy Cao
- Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Russell T Shinohara
- Center for Data Science and AI for Integrated Diagnostics (AI2D), and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Penn Statistics in Imaging and Visualization Center, and Center for Clinical Epidemiology ce and AI for Integrated Diagnostics (AI2D), and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ujjwal Baid
- Department of Pathology & Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Alexander Getka
- Center for Data Science and AI for Integrated Diagnostics (AI2D), and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Penn Statistics in Imaging and Visualization Center, and Center for Clinical Epidemiology ce and AI for Integrated Diagnostics (AI2D), and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sarthak Pati
- Department of Pathology & Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Ashish Singh
- Center for Data Science and AI for Integrated Diagnostics (AI2D), and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Penn Statistics in Imaging and Visualization Center, and Center for Clinical Epidemiology ce and AI for Integrated Diagnostics (AI2D), and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Evan Calabrese
- Department of Radiology, Duke University, Durham, North Carolina, USA
| | - Susan Chang
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, USA
| | - Jeffrey Rudie
- Department of Radiology, University of California San Diego, San Diego, California, USA
| | - Aristeidis Sotiras
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Pamela LaMontagne
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Daniel S Marcus
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Mikhail Milchenko
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Arash Nazeri
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Carmen Balana
- B-ARGO Group, Institut Investigació Germans Trias i Pujol (IGTP), Badalona (Barcelona), Catalonia, Spain
| | - Jaume Capellades
- Research Unit (IDIR), Image Diagnosis Institute, Badalona, Spain
| | - Josep Puig
- Department of Radiology (CDI), Hospital Clínic and IDIBAPS, Barcelona, Spain
| | - Chaitra Badve
- Department of Radiology, Case Western Reserve University and University Hospitals of Cleveland, Cleveland, Ohio, USA
| | - Jill S Barnholtz-Sloan
- Trans-Divisional Research Program (TDRP), Division of Cancer Epidemiology and Genetics (DCEG), National Cancer Institute, Bethesda, Maryland, USA
- Central Brain Tumor Registry of the United States, Hinsdale, Illinois, USA
- Center for Biomedical Informatics and Information Technology (CBIIT), National Cancer Institute, Bethesda, Maryland, USA
| | - Andrew E Sloan
- Brain and Tumor Neurosurgery, Neurosurgical Oncology, Piedmont Health, Atlanta, Georgia, USA
- Seidman Cancer Center and Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, USA
| | - Vachan Vadmal
- Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Kristin Waite
- Center for Biomedical Informatics and Information Technology (CBIIT), National Cancer Institute, Bethesda, Maryland, USA
- Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Murat Ak
- Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Rivka R Colen
- Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Yae Won Park
- Department of Radiology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sung Soo Ahn
- Department of Radiology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jong Hee Chang
- Department of Neurosurgery, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Tumor Center, Severance Hospital, Yonsei University Health System, Seoul, Republic of Korea
| | - Yoon Seong Choi
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Clinical Imaging Research Centre, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Seung-Koo Lee
- Department of Radiology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Gregory S Alexander
- Department of Radiation Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Ayesha S Ali
- Department of Radiation Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Adam P Dicker
- Department of Radiation Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Adam E Flanders
- Department of Radiology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Spencer Liem
- Department of Radiation Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Joseph Lombardo
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
- Department of Radiation Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Wenyin Shi
- Department of Radiation Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Gaurav Shukla
- Center for Data Science and AI for Integrated Diagnostics (AI2D), and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Penn Statistics in Imaging and Visualization Center, and Center for Clinical Epidemiology ce and AI for Integrated Diagnostics (AI2D), and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Radiation Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
- Department of Radiation Oncology, Christiana Care Health System, Philadelphia, Pennsylvania, USA
| | - Brent Griffith
- Department of Radiology, Henry Ford Health System, Detroit, Michigan, USA
| | - Laila M Poisson
- Department of Public Health Sciences, Center for Bioinformatics, Henry Ford Health System, Detroit, Michigan, USA
- Department of Neurosurgery, Hermelin Brain Tumor Center, Henry Ford Cancer Institute, Henry Ford Health, Detroit, Michigan, USA
| | - Lisa R Rogers
- Department of Neurosurgery, Hermelin Brain Tumor Center, Henry Ford Cancer Institute, Henry Ford Health, Detroit, Michigan, USA
| | | | - Thomas C Booth
- School of Biomedical Engineering and Imaging Sciences, King’s College London, London, UK
- Department of Neuroradiology, Ruskin Wing, King’s College Hospital NHS Foundation Trust, London, UK
| | - Rajan Jain
- Department of Radiology, New York University Langone Health, New York, New York, USA
- Department of Neurosurgery, New York University Langone Health, New York, New York, USA
| | - Matthew Lee
- Department of Radiology, New York University Langone Health, New York, New York, USA
| | - Abhishek Mahajan
- Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
- The Clatterbridge Cancer Centre NHS Foundation Trust, Liverpool, UK
| | - Arnab Chakravarti
- Department of Radiation Oncology, The James Cancer Hospital at the Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Joshua D Palmer
- Department of Radiation Oncology, The James Cancer Hospital at the Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Dominic DiCostanzo
- Department of Radiation Oncology, The James Cancer Hospital at the Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | | | - Santiago Cepeda
- Department of Neurosurgery, University Hospital Río Hortega, Valladolid, Spain
| | - Orazio Santo Santonocito
- Division of Neurosurgery, Spedali Riuniti di Livorno-Azienda USL Toscana Nord-Ovest, Livorno, Italy
| | - Anna Luisa Di Stefano
- Division of Neurosurgery, Spedali Riuniti di Livorno-Azienda USL Toscana Nord-Ovest, Livorno, Italy
| | - Benedikt Wiestler
- Department of Neuroradiology, Technical University of Munich, Munchen, Germany
| | - Elias R Melhem
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Graeme F Woodworth
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Pallavi Tiwari
- Department of Radiology, University of Wisconsin, Madison, Wisconsin, USA
- Department of Biomedical Engineering, University of Wisconsin, Madison, Wisconsin, USA
| | - Pablo Valdes
- Department of Neurosurgery, University of Texas Medical Branch, Galveston, Texas, USA
| | - Yuji Matsumoto
- Department of Neurological Surgery, Okayama University, Okayama, Japan
| | - Yoshihiro Otani
- Department of Neurological Surgery, Okayama University, Okayama, Japan
| | - Ryoji Imoto
- Department of Neurological Surgery, Okayama University, Okayama, Japan
| | - Mariam Aboian
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Shinichiro Koizumi
- Department of Neurosurgery, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Kazuhiko Kurozumi
- Department of Neurosurgery, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Toru Kawakatsu
- Department of Neurosurgery, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Kimberley Alexander
- Department of Neurosurgery, Chris O’Brien Lifehouse, Camperdown, Australia
- Faculty of Medicine and Health, University of Sydney, Camperdown, Australia
- Department of Neuropathology, Royal Prince Alfred Hospital, Camperdown, Australia
| | - Laveniya Satgunaseelan
- Department of Neurosurgery, Chris O’Brien Lifehouse, Camperdown, Australia
- Faculty of Medicine and Health, University of Sydney, Camperdown, Australia
- Department of Neuropathology, Royal Prince Alfred Hospital, Camperdown, Australia
| | - Aaron M Rulseh
- Department of Radiology, Na Homolce Hospital, Prague, Czechia
| | - Stephen J Bagley
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- GBM Translational Center of Excellence, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Michel Bilello
- Center for Data Science and AI for Integrated Diagnostics (AI2D), and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Penn Statistics in Imaging and Visualization Center, and Center for Clinical Epidemiology ce and AI for Integrated Diagnostics (AI2D), and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Zev A Binder
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- GBM Translational Center of Excellence, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Steven Brem
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- GBM Translational Center of Excellence, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Arati S Desai
- GBM Translational Center of Excellence, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Robert A Lustig
- Department of Radiation-Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Eileen Maloney
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Timothy Prior
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Nduka Amankulor
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- GBM Translational Center of Excellence, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - MacLean P Nasrallah
- Center for Data Science and AI for Integrated Diagnostics (AI2D), and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Penn Statistics in Imaging and Visualization Center, and Center for Clinical Epidemiology ce and AI for Integrated Diagnostics (AI2D), and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, Pennsylvania, USA
- GBM Translational Center of Excellence, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Pathology & Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Donald M O’Rourke
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- GBM Translational Center of Excellence, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Suyash Mohan
- Center for Data Science and AI for Integrated Diagnostics (AI2D), and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Penn Statistics in Imaging and Visualization Center, and Center for Clinical Epidemiology ce and AI for Integrated Diagnostics (AI2D), and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Christos Davatzikos
- Center for Data Science and AI for Integrated Diagnostics (AI2D), and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Penn Statistics in Imaging and Visualization Center, and Center for Clinical Epidemiology ce and AI for Integrated Diagnostics (AI2D), and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
2
|
Yoshimura H, Kawahara D, Saito A, Ozawa S, Nagata Y. Prediction of prognosis in glioblastoma with radiomics features extracted by synthetic MRI images using cycle-consistent GAN. Phys Eng Sci Med 2024; 47:1227-1243. [PMID: 38884673 PMCID: PMC11408565 DOI: 10.1007/s13246-024-01443-8] [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: 05/24/2023] [Accepted: 05/14/2024] [Indexed: 06/18/2024]
Abstract
To propose a style transfer model for multi-contrast magnetic resonance imaging (MRI) images with a cycle-consistent generative adversarial network (CycleGAN) and evaluate the image quality and prognosis prediction performance for glioblastoma (GBM) patients from the extracted radiomics features. Style transfer models of T1 weighted MRI image (T1w) to T2 weighted MRI image (T2w) and T2w to T1w with CycleGAN were constructed using the BraTS dataset. The style transfer model was validated with the Cancer Genome Atlas Glioblastoma Multiforme (TCGA-GBM) dataset. Moreover, imaging features were extracted from real and synthesized images. These features were transformed to rad-scores by the least absolute shrinkage and selection operator (LASSO)-Cox regression. The prognosis performance was estimated by the Kaplan-Meier method. For the accuracy of the image quality of the real and synthesized MRI images, the MI, RMSE, PSNR, and SSIM were 0.991 ± 2.10 × 10 - 4 , 2.79 ± 0.16, 40.16 ± 0.38, and 0.995 ± 2.11 × 10 - 4 , for T2w, and .992 ± 2.63 × 10 - 4 , 2.49 ± 6.89 × 10 - 2 , 40.51 ± 0.22, and 0.993 ± 3.40 × 10 - 4 for T1w, respectively. The survival time had a significant difference between good and poor prognosis groups for both real and synthesized T2w (p < 0.05). However, the survival time had no significant difference between good and poor prognosis groups for both real and synthesized T1w. On the other hand, there was no significant difference between the real and synthesized T2w in both good and poor prognoses. The results of T1w were similar in the point that there was no significant difference between the real and synthesized T1w. It was found that the synthesized image could be used for prognosis prediction. The proposed prognostic model using CycleGAN could reduce the cost and time of image scanning, leading to a promotion to build the patient's outcome prediction with multi-contrast images.
Collapse
Affiliation(s)
- Hisanori Yoshimura
- Department of Radiation Oncology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, 734-8551, Japan
- Department of Radiology, National Hospital Organization Kure Medical Center, Hiroshima, Japan
| | - Daisuke Kawahara
- Department of Radiation Oncology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, 734-8551, Japan.
| | - Akito Saito
- Department of Radiation Oncology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, 734-8551, Japan
| | - Shuichi Ozawa
- Department of Radiation Oncology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, 734-8551, Japan
- Hiroshima High-Precision Radiotherapy Cancer Center, Hiroshima, 732-0057, Japan
| | - Yasushi Nagata
- Department of Radiation Oncology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, 734-8551, Japan
- Hiroshima High-Precision Radiotherapy Cancer Center, Hiroshima, 732-0057, Japan
| |
Collapse
|
3
|
Fatania K, Frood R, Mistry H, Short SC, O’Connor J, Scarsbrook AF, Currie S. Tumour Size and Overall Survival in a Cohort of Patients with Unifocal Glioblastoma: A Uni- and Multivariable Prognostic Modelling and Resampling Study. Cancers (Basel) 2024; 16:1301. [PMID: 38610979 PMCID: PMC11011077 DOI: 10.3390/cancers16071301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 03/15/2024] [Accepted: 03/20/2024] [Indexed: 04/14/2024] Open
Abstract
Published models inconsistently associate glioblastoma size with overall survival (OS). This study aimed to investigate the prognostic effect of tumour size in a large cohort of patients diagnosed with GBM and interrogate how sample size and non-linear transformations may impact on the likelihood of finding a prognostic effect. In total, 279 patients with a IDH-wildtype unifocal WHO grade 4 GBM between 2014 and 2020 from a retrospective cohort were included. Uni-/multivariable association between core volume, whole volume (CV and WV), and diameter with OS was assessed with (1) Cox proportional hazard models +/- log transformation and (2) resampling with 1,000,000 repetitions and varying sample size to identify the percentage of models, which showed a significant effect of tumour size. Models adjusted for operation type and a diameter model adjusted for all clinical variables remained significant (p = 0.03). Multivariable resampling increased the significant effects (p < 0.05) of all size variables as sample size increased. Log transformation also had a large effect on the chances of a prognostic effect of WV. For models adjusted for operation type, 19.5% of WV vs. 26.3% log-WV (n = 50) and 69.9% WV and 89.9% log-WV (n = 279) were significant. In this large well-curated cohort, multivariable modelling and resampling suggest tumour volume is prognostic at larger sample sizes and with log transformation for WV.
Collapse
Affiliation(s)
- Kavi Fatania
- Department of Radiology, Leeds Teaching Hospitals NHS Trust, Leeds General Infirmary, Leeds LS1 3EX, UK (A.F.S.); (S.C.)
- Leeds Institute of Medical Research, University of Leeds, Leeds LS2 9TJ, UK;
| | - Russell Frood
- Department of Radiology, Leeds Teaching Hospitals NHS Trust, Leeds General Infirmary, Leeds LS1 3EX, UK (A.F.S.); (S.C.)
- Leeds Institute of Medical Research, University of Leeds, Leeds LS2 9TJ, UK;
| | - Hitesh Mistry
- Division of Cancer Sciences, The University of Manchester, Manchester M13 9PL, UK; (H.M.)
| | - Susan C. Short
- Leeds Institute of Medical Research, University of Leeds, Leeds LS2 9TJ, UK;
- Department of Oncology, Leeds Teaching Hospitals NHS Trust, St James’s University Hospital, Leeds LS9 7TF, UK
| | - James O’Connor
- Division of Cancer Sciences, The University of Manchester, Manchester M13 9PL, UK; (H.M.)
- Department of Radiology, The Christie Hospital, Manchester M20 4BX, UK
- Division of Radiotherapy and Imaging, Institute of Cancer Research, London SM2 5NG, UK
| | - Andrew F. Scarsbrook
- Department of Radiology, Leeds Teaching Hospitals NHS Trust, Leeds General Infirmary, Leeds LS1 3EX, UK (A.F.S.); (S.C.)
- Leeds Institute of Medical Research, University of Leeds, Leeds LS2 9TJ, UK;
| | - Stuart Currie
- Department of Radiology, Leeds Teaching Hospitals NHS Trust, Leeds General Infirmary, Leeds LS1 3EX, UK (A.F.S.); (S.C.)
- Leeds Institute of Medical Research, University of Leeds, Leeds LS2 9TJ, UK;
| |
Collapse
|
4
|
Guo J, Fathi Kazerooni A, Toorens E, Akbari H, Yu F, Sako C, Mamourian E, Shinohara RT, Koumenis C, Bagley SJ, Morrissette JJD, Binder ZA, Brem S, Mohan S, Lustig RA, O'Rourke DM, Ganguly T, Bakas S, Nasrallah MP, Davatzikos C. Integrating imaging and genomic data for the discovery of distinct glioblastoma subtypes: a joint learning approach. Sci Rep 2024; 14:4922. [PMID: 38418494 PMCID: PMC10902376 DOI: 10.1038/s41598-024-55072-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 02/19/2024] [Indexed: 03/01/2024] Open
Abstract
Glioblastoma is a highly heterogeneous disease, with variations observed at both phenotypical and molecular levels. Personalized therapies would be facilitated by non-invasive in vivo approaches for characterizing this heterogeneity. In this study, we developed unsupervised joint machine learning between radiomic and genomic data, thereby identifying distinct glioblastoma subtypes. A retrospective cohort of 571 IDH-wildtype glioblastoma patients were included in the study, and pre-operative multi-parametric MRI scans and targeted next-generation sequencing (NGS) data were collected. L21-norm minimization was used to select a subset of 12 radiomic features from the MRI scans, and 13 key driver genes from the five main signal pathways most affected in glioblastoma were selected from the genomic data. Subtypes were identified using a joint learning approach called Anchor-based Partial Multi-modal Clustering on both radiomic and genomic modalities. Kaplan-Meier analysis identified three distinct glioblastoma subtypes: high-risk, medium-risk, and low-risk, based on overall survival outcome (p < 0.05, log-rank test; Hazard Ratio = 1.64, 95% CI 1.17-2.31, Cox proportional hazard model on high-risk and low-risk subtypes). The three subtypes displayed different phenotypical and molecular characteristics in terms of imaging histogram, co-occurrence of genes, and correlation between the two modalities. Our findings demonstrate the synergistic value of integrated radiomic signatures and molecular characteristics for glioblastoma subtyping. Joint learning on both modalities can aid in better understanding the molecular basis of phenotypical signatures of glioblastoma, and provide insights into the biological underpinnings of tumor formation and progression.
Collapse
Affiliation(s)
- Jun Guo
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, 3700 Hamilton Walk, 7Th Floor, Philadelphia, PA, 19104, USA
- Center for AI and Data Science for Integrated Diagnostics, University of Pennsylvania, Philadelphia, USA
- 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, 3700 Hamilton Walk, 7Th Floor, Philadelphia, PA, 19104, USA
- Center for AI and Data Science for Integrated Diagnostics, University of Pennsylvania, Philadelphia, USA
- Center for Data-Driven Discovery in Biomedicine (D3b), Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Erik Toorens
- Penn Genomic Analysis Core, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hamed Akbari
- Department of Bioengineering, School of Engineering, Santa Clara University, Santa Clara, CA, USA
| | - Fanyang Yu
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, 3700 Hamilton Walk, 7Th Floor, Philadelphia, PA, 19104, USA
- Center for AI and Data Science for Integrated Diagnostics, University of Pennsylvania, Philadelphia, USA
| | - Chiharu Sako
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, 3700 Hamilton Walk, 7Th Floor, Philadelphia, PA, 19104, USA
- Center for AI and Data Science for Integrated Diagnostics, University of Pennsylvania, Philadelphia, 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, 3700 Hamilton Walk, 7Th Floor, Philadelphia, PA, 19104, 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, 3700 Hamilton Walk, 7Th Floor, Philadelphia, PA, 19104, USA
- Penn Statistics in Imaging and Visualization (PennSIVE) Center, Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Constantinos Koumenis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Stephen J Bagley
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Jennifer J D Morrissette
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zev A Binder
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Steven Brem
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Suyash Mohan
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, 3700 Hamilton Walk, 7Th Floor, Philadelphia, PA, 19104, USA
- Center for AI and Data Science for Integrated Diagnostics, University of Pennsylvania, Philadelphia, 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
- Abramson Cancer Center, 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
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Tapan Ganguly
- Penn Genomic Analysis Core, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Spyridon Bakas
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, 3700 Hamilton Walk, 7Th Floor, Philadelphia, PA, 19104, USA
- Center for AI and Data Science for Integrated Diagnostics, University of Pennsylvania, Philadelphia, 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
- Division of Computational Pathology, Department of Pathology & Laboratory Medicine, School of Medicine, Indiana University, Indianapolis, IN, USA
| | - MacLean P Nasrallah
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, 3700 Hamilton Walk, 7Th Floor, Philadelphia, PA, 19104, USA
- Center for AI and Data Science for Integrated Diagnostics, University of Pennsylvania, Philadelphia, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christos Davatzikos
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, 3700 Hamilton Walk, 7Th Floor, Philadelphia, PA, 19104, USA.
- Center for AI and Data Science for Integrated Diagnostics, University of Pennsylvania, Philadelphia, USA.
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
5
|
Rathore S, Iftikhar MA, Chaddad A, Singh A, Gillani Z, Abdulkadir A. Imaging phenotypes predict overall survival in glioma more accurate than basic demographic and cell mutation profiles. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2023; 242:107812. [PMID: 37757566 DOI: 10.1016/j.cmpb.2023.107812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 05/14/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023]
Abstract
BACKGROUND Magnetic resonance imaging (MRI), digital pathology imaging (PATH), demographics, and IDH mutation status predict overall survival (OS) in glioma. Identifying and characterizing predictive features in the different modalities may improve OS prediction accuracy. PURPOSE To evaluate the OS prediction accuracy of combinations of prognostic markers in glioma patients. MATERIALS AND METHODS Multi-contrast MRI, comprising T1-weighted, T1-weighted post-contrast, T2-weighted, T2 fluid-attenuated-inversion-recovery, and pathology images from glioma patients (n = 160) were retrospectively collected (1983-2008) from TCGA alongside age and sex. Phenotypic profiling of tumors was performed by quantifying the radiographic and histopathologic descriptors extracted from the delineated region-of-interest in MRI and PATH images. A Cox proportional hazard model was trained with the MRI and PATH features, IDH mutation status, and basic demographic variables (age and sex) to predict OS. The performance was evaluated in a split-train-test configuration using the concordance-index, computed between the predicted risk score and observed OS. RESULTS The average age of patients was 51.2years (women: n = 77, age-range=18-84years; men: n = 83, age-range=21-80years). The median OS of the participants was 494.5 (range,3-4752), 481 (range,7-4752), and 524.5 days (range,3-2869), respectively, in complete dataset, training, and test datasets. The addition of MRI or PATH features improved prediction of OS when compared to models based on age, sex, and mutation status alone or their combination (p < 0.001). The full multi-omics model integrated MRI, PATH, clinical, and genetic profiles and predicted the OS best (c-index= 0.87). CONCLUSION The combination of imaging, genetic, and clinical profiles leads to a more accurate prognosis than the clinical and/or mutation status.
Collapse
Affiliation(s)
- Saima Rathore
- AVID Radiopharmaceuticals, Philadelphia, PA, USA; Eli Lilly and Company, Indianapolis, IN, USA.
| | | | - Ahmad Chaddad
- School of Artificial Intelligence, GUET, Guilin, China
| | - Ashish Singh
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Zeeshan Gillani
- Comsats University Islamabad, Lahore Campus, Lahore, Pakistan
| | - Ahmed Abdulkadir
- Center for Research in Neurosciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland; Center for Artificial Intelligence, Zurich University of Applied Sciences, Winterthur, ZH, Switzerland
| |
Collapse
|
6
|
Lau KS, Ruisi I, Back M. Association of MRI Volume Parameters in Predicting Patient Outcome at Time of Initial Diagnosis of Glioblastoma. Brain Sci 2023; 13:1579. [PMID: 38002539 PMCID: PMC10670247 DOI: 10.3390/brainsci13111579] [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: 09/29/2023] [Revised: 11/08/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
PURPOSE Patients with glioblastoma (GBM) may demonstrate varying patterns of infiltration and relapse. Improving the ability to predict these patterns may influence the management strategies at the time of initial diagnosis. This study aims to examine the impact of the ratio (T2/T1) of the non-enhancing volume in T2-weighted images (T2) to the enhancing volume in MRI T1-weighted gadolinium-enhanced images (T1gad) on patient outcome. METHODS AND MATERIALS A retrospective audit was performed from established prospective databases in patients managed consecutively with radiation therapy (RT) for GBM between 2016 and 2019. Patient, tumour and treatment-related factors were assessed in relation to outcome. Volumetric data from the initial diagnostic MRI were obtained via the manual segmentation of the T1gd and T2 abnormalities. A T2/T1 ratio was calculated from these volumes. The initial relapse site was assessed on MRI in relation to the site of the original T1gad volume and surgical cavity. The major endpoints were median relapse-free survival (RFS) from the date of diagnosis and site of initial relapse (defined as either local at the initial surgical site or any distance more than 20 mm from initial T1gad abnormality). The analysis was performed for association between known prognostic factors as well as the radiological factors using log-rank tests for subgroup comparisons, with correction for multiple comparisons. RESULTS One hundred and seventy-seven patients with GBM were managed consecutively with RT between 2016 and 2019 and were eligible for the analysis. The median age was 62 years. Seventy-four percent were managed under a 60Gy (Stupp) protocol, whilst 26% were on a 40Gy (Elderly) protocol. Major neuroanatomical subsites were Lateral Temporal (18%), Anterior Temporal (13%) and Medial Frontal (10%). Median volumes on T1gd and T2 were 20 cm3 (q1-3:8-43) and 37 cm3 (q1-3: 17-70), respectively. The median T2/T1 ratio was 2.1. For the whole cohort, the median OS was 16.0 months (95%CI:14.1-18.0). One hundred and forty-eight patients have relapsed with a median RFS of 11.4 months (95%CI:10.4-12.5). A component of distant relapse was evident in 43.9% of relapses, with 23.6% isolated relapse. Better ECOG performance Status (p = 0.007), greater extent of resection (p = 0.020), MGMT methylation (p < 0.001) and RT60Gy Dose (p = 0.050) were associated with improved RFS. Although the continuous variable of initial T1gd volume (p = 0.39) and T2 volume (p = 0.23) were not associated with RFS, the lowest T2/T1 quartile (reflecting a relatively lower T2 volume compared to T1gd volume) was significantly associated with improved RFS (p = 0.016) compared with the highest quartile. The lowest T2/T1 ratio quartile was also associated with a lower risk of distant relapse (p = 0.031). CONCLUSION In patients diagnosed with GBM, the volumetric parameters of the diagnostic MRI with a ratio of T2 and T1gad abnormality may assist in the prediction of relapse-free survival and patterns of relapse. A further understanding of these relationships has the potential to impact the design of future radiation therapy target volume delineation protocols.
Collapse
Affiliation(s)
- Kin Sing Lau
- Department of Radiation Oncology, Northern Sydney Cancer Centre, Royal North Shore Hospital, Sydney, NSW 2065, Australia;
- Central Coast Cancer Centre, Gosford Hospital, Gosford, NSW 2250, Australia
| | - Isidoro Ruisi
- Central Coast Cancer Centre, Gosford Hospital, Gosford, NSW 2250, Australia
| | - Michael Back
- Department of Radiation Oncology, Northern Sydney Cancer Centre, Royal North Shore Hospital, Sydney, NSW 2065, Australia;
- Central Coast Cancer Centre, Gosford Hospital, Gosford, NSW 2250, Australia
- Genesis Care, Sydney, NSW 2015, Australia
- Sydney Medical School, University of Sydney, Sydney, NSW 2050, Australia
- The Brain Cancer Group, Sydney, NSW 2065, Australia
| |
Collapse
|
7
|
Shah AS, Sylvester PT, Yahanda AT, Vellimana AK, Dunn GP, Evans J, Rich KM, Dowling JL, Leuthardt EC, Dacey RG, Kim AH, Grubb RL, Zipfel GJ, Oswood M, Jensen RL, Sutherland GR, Cahill DP, Abram SR, Honeycutt J, Shah M, Tao Y, Chicoine MR. Intraoperative MRI for newly diagnosed supratentorial glioblastoma: a multicenter-registry comparative study to conventional surgery. J Neurosurg 2021; 135:505-514. [PMID: 33035996 DOI: 10.3171/2020.6.jns19287] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 06/04/2020] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Intraoperative MRI (iMRI) is used in the surgical treatment of glioblastoma, with uncertain effects on outcomes. The authors evaluated the impact of iMRI on extent of resection (EOR) and overall survival (OS) while controlling for other known and suspected predictors. METHODS A multicenter retrospective cohort of 640 adult patients with newly diagnosed supratentorial glioblastoma who underwent resection was evaluated. iMRI was performed in 332/640 cases (51.9%). Reviews of MRI features and tumor volumetric analysis were performed on a subsample of cases (n = 286; 110 non-iMRI, 176 iMRI) from a single institution. RESULTS The median age was 60.0 years (mean 58.5 years, range 20.5-86.3 years). The median OS was 17.0 months (95% CI 15.6-18.4 months). Gross-total resection (GTR) was achieved in 403/640 cases (63.0%). Kaplan-Meier analysis of 286 cases with volumetric analysis for EOR (grouped into 100%, 95%-99%, 80%-94%, and 50%-79%) showed longer OS for 100% EOR compared to all other groups (p < 0.01). Additional resection after iMRI was performed in 104/122 cases (85.2%) with initial subtotal resection (STR), leading to a 6.3% mean increase in EOR and a 2.2-cm3 mean decrease in tumor volume. For iMRI cases with volumetric analysis, the GTR rate increased from 54/176 (30.7%) on iMRI to 126/176 (71.5%) postoperatively. The EOR was significantly higher in the iMRI group for intended GTR and STR groups (p = 0.02 and p < 0.01, respectively). Predictors of GTR on multivariate logistic regression included iMRI use and intended GTR. Predictors of shorter OS on multivariate Cox regression included older age, STR, isocitrate dehydrogenase 1 (IDH1) wild type, no O 6-methylguanine DNA methyltransferase (MGMT) methylation, and no Stupp therapy. iMRI was a significant predictor of OS on univariate (HR 0.82, 95% CI 0.69-0.98; p = 0.03) but not multivariate analyses. Use of iMRI was not associated with an increased rate of new permanent neurological deficits. CONCLUSIONS GTR increased OS for patients with newly diagnosed glioblastoma after adjusting for other prognostic factors. iMRI increased EOR and GTR rate and was a significant predictor of GTR on multivariate analysis; however, iMRI was not an independent predictor of OS. Additional supporting evidence is needed to determine the clinical benefit of iMRI in the management of glioblastoma.
Collapse
Affiliation(s)
- Amar S Shah
- 1Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri
| | - Peter T Sylvester
- 1Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri
| | - Alexander T Yahanda
- 1Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri
| | - Ananth K Vellimana
- 1Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri
| | - Gavin P Dunn
- 1Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri
| | - John Evans
- 1Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri
| | - Keith M Rich
- 1Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri
| | - Joshua L Dowling
- 1Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri
| | - Eric C Leuthardt
- 1Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri
| | - Ralph G Dacey
- 1Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri
| | - Albert H Kim
- 1Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri
| | - Robert L Grubb
- 1Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri
| | - Gregory J Zipfel
- 1Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri
| | - Mark Oswood
- 2Department of Radiology, University of Minnesota, Minneapolis, Minnesota
- 3Allina Health, Minneapolis, Minnesota
| | - Randy L Jensen
- 4Department of Neurosurgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Garnette R Sutherland
- 5Department of Clinical Sciences and Hotchkiss Brain Institute, University of Calgary, Alberta, Canada
| | - Daniel P Cahill
- 6Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts
| | - Steven R Abram
- 7Department of Neurosurgery, St. Thomas Hospital, Nashville, Tennessee
| | - John Honeycutt
- 8Department of Neurosurgery, Cook Children's Hospital, Fort Worth, Texas; and
| | - Mitesh Shah
- 9Department of Neurological Surgery, Goodman Campbell and Indiana University, Indianapolis, Indiana
| | - Yu Tao
- 1Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri
| | - Michael R Chicoine
- 1Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
8
|
Soltani M, Bonakdar A, Shakourifar N, Babaei R, Raahemifar K. Efficacy of Location-Based Features for Survival Prediction of Patients With Glioblastoma Depending on Resection Status. Front Oncol 2021; 11:661123. [PMID: 34295809 PMCID: PMC8290179 DOI: 10.3389/fonc.2021.661123] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 06/14/2021] [Indexed: 12/24/2022] Open
Abstract
Cancer stands out as one of the fatal diseases people are facing all the time. Each year, a countless number of people die because of the late diagnosis of cancer or wrong treatments. Glioma, one of the most common primary brain tumors, has different aggressiveness and sub-regions, which can affect the risk of disease. Although prediction of overall survival based on multimodal magnetic resonance imaging (MRI) is challenging, in this study, we assess if and how location-based features of tumors can affect overall survival prediction. This approach is evaluated independently and in combination with radiomic features. The process is carried out on a data set entailing MRI images of patients with glioblastoma. To assess the impact of resection status, the data set is divided into two groups, patients were reported as gross total resection and unknown resection status. Then, different machine learning algorithms were used to evaluate how location features are linked with overall survival. Results from regression models indicate that location-based features have considerable effects on the patients' overall survival independently. Additionally, classifier models show an improvement in prediction accuracy by the addition of location-based features to radiomic features.
Collapse
Affiliation(s)
- Madjid Soltani
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
- Department of Electrical and Computer Engineering, University of Waterloo, Waterloo, ON, Canada
- Centre for Biotechnology and Bioengineering (CBB), University of Waterloo, Waterloo, ON, Canada
- Advanced Bioengineering Initiative Center, Computational Medicine Center, K. N. Toosi University of Technology, Tehran, Iran
| | - Armin Bonakdar
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| | - Nastaran Shakourifar
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| | - Reza Babaei
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| | - Kaamran Raahemifar
- College of Information Sciences and Technology (IST), Data Science and Artificial Intelligence Program, Penn State University, State College, Pennsylvania, PA, United States
- Chemical Engineering Department, University of Waterloo, Waterloo, ON, Canada
- Optometry & Vision Science Department, University of Waterloo, Waterloo, ON, Canada
| |
Collapse
|
9
|
18F-FET PET Uptake Characteristics of Long-Term IDH-Wildtype Diffuse Glioma Survivors. Cancers (Basel) 2021; 13:cancers13133163. [PMID: 34202726 PMCID: PMC8268019 DOI: 10.3390/cancers13133163] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/08/2021] [Accepted: 06/18/2021] [Indexed: 11/28/2022] Open
Abstract
Simple Summary IDH-wildtype (IDHwt) gliomas represent a tumor entity with poor overall survival. Only rare cases have an overall survival over several years. Dynamic and static 18F-FET PET is recommended as valuable complementary tool for glioma imaging in gliomas. This study shows that, besides molecular genetic prognosticators, long survival (≥36 months survival) in IDHwt gliomas is associated with a longer time-to-peak and smaller volume on 18F-FET PET at initial diagnosis compared to glioma patients with a short-term survival (≤15 months survival). 18F-FET uptake intensity and MRI-derived tumor size do not differ in patients with long-term survival compared to patient with a short-term survival. Abstract Background: IDHwt diffuse gliomas represent the tumor entity with one of the worst clinical outcomes. Only rare cases present with a long-term survival of several years. Here we aimed at comparing the uptake characteristics on dynamic 18F-FET PET, clinical and molecular genetic parameters of long-term survivors (LTS) versus short-term survivors (STS): Methods: Patients with de-novo IDHwt glioma (WHO grade III/IV) and 18F-FET PET prior to any therapy were stratified into LTS (≥36 months survival) and STS (≤15 months survival). Static and dynamic 18F-FET PET parameters (mean/maximal tumor-to-background ratio (TBRmean/max), biological tumor volume (BTV), minimal time-to-peak (TTPmin)), diameter and volume of contrast-enhancement on MRI, clinical parameters (age, sex, Karnofksy-performance-score), mode of surgery; initial treatment and molecular genetics were assessed and compared between LTS and STS. Results: Overall, 75 IDHwt glioma patients were included (26 LTS, 49 STS). LTS were significantly younger (p < 0.001), had a higher rate of WHO grade III glioma (p = 0.032), of O(6)-Methylguanine-DNA methyltransferase (MGMT) promoter methylation (p < 0.001) and missing Telomerase reverse transcriptase promoter (TERTp) mutations (p = 0.004) compared to STS. On imaging, LTS showed a smaller median BTV (p = 0.017) and a significantly longer TTPmin (p = 0.008) on 18F-FET PET than STS, while uptake intensity (TBRmean/max) did not differ. In contrast to the tumor-volume on PET, MRI-derived parameters such as tumor size as well as all other above-mentioned parameters did not differ between LTS and STS (p > 0.05 each). Conclusion: Besides molecular genetic prognosticators, a long survival time in IDHwt glioma patients is associated with a longer TTPmin as well as a smaller BTV on 18F-FET PET at initial diagnosis. 18F-FET uptake intensity as well as the MRI-derived tumor size (volume and maximal diameter) do not differ in patients with long-term survival.
Collapse
|
10
|
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
|
11
|
Li M, Ren X, Dong G, Wang J, Jiang H, Yang C, Zhao X, Zhu Q, Cui Y, Yu K, Lin S. Distinguishing Pseudoprogression From True Early Progression in Isocitrate Dehydrogenase Wild-Type Glioblastoma by Interrogating Clinical, Radiological, and Molecular Features. Front Oncol 2021; 11:627325. [PMID: 33959496 PMCID: PMC8093388 DOI: 10.3389/fonc.2021.627325] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 02/12/2021] [Indexed: 12/03/2022] Open
Abstract
Background: Pseudoprogression (PsP) mimics true early progression (TeP) in conventional imaging, which poses a diagnostic challenge in glioblastoma (GBM) patients who undergo standard concurrent chemoradiation (CCRT). This study aimed to investigate whether perioperative markers could distinguish and predict PsP from TeP in de novo isocitrate dehydrogenase (IDH) wild-type GBM patients. Methods: New or progressive gadolinium-enhancing lesions that emerged within 12 weeks after CCRT were defined as early progression. Lesions that remained stable or spontaneously regressed were classified as PsP, otherwise persistently enlarged as TeP. Clinical, radiological, and molecular information were collected for further analysis. Patients in the early progression subgroup were divided into derivation and validation sets (7:3, according to operation date). Results: Among 234 consecutive cases enrolled in this retrospective study, the incidences of PsP, TeP, and neither patterns of progression (nP) were 26.1% (61/234), 37.6% (88/234), and 36.3% (85/234), respectively. In the early progression subgroup, univariate analysis demonstrated female (OR: 2.161, P = 0.026), gross total removal (GTR) of the tumor (OR: 6.571, P < 001), located in the frontal lobe (OR: 2.561, P = 0.008), non-subventricular zone (SVZ) infringement (OR: 10.937, P < 0.001), and methylated O-6-methylguanine-DNA methyltransferase (MGMT) promoter (mMGMTp) (OR: 9.737, P < 0.001) were correlated with PsP, while GTR, non-SVZ infringement, and mMGMTp were further validated in multivariate analysis. Integrating quantitative MGMTp methylation levels from pyrosequencing, GTR, and non-SVZ infringement showed the best discriminative ability in the random forest model for derivation and validation set (AUC: 0.937, 0.911, respectively). Furthermore, a nomogram could effectively evaluate the importance of those markers in developing PsP (C-index: 0.916) and had a well-fitted calibration curve. Conclusion: Integrating those clinical, radiological, and molecular features provided a novel and robust method to distinguish PsP from TeP, which was crucial for subsequent clinical decision making, clinical trial enrollment, and prognostic assessment. By in-depth interrogation of perioperative markers, clinicians could distinguish PsP from TeP independent from advanced imaging.
Collapse
Affiliation(s)
- Mingxiao Li
- Department of Neurosurgery, National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xiaohui Ren
- Department of Neurosurgery, National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Gehong Dong
- Department of Pathology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jincheng Wang
- Department of Radiology, Peking University Cancer Hospital, Beijing, China
| | - Haihui Jiang
- Department of Neurosurgery, National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Chuanwei Yang
- Department of Neurosurgery, National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xuzhe Zhao
- Department of Neurosurgery, National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Qinghui Zhu
- Department of Neurosurgery, National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yong Cui
- Department of Neurosurgery, National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Kefu Yu
- Department of Pharmacy, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Song Lin
- Department of Neurosurgery, National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Brain Tumor, Center of Brain Tumor, Institute for Brain Disorders, Beijing, China
| |
Collapse
|
12
|
Xu Y, He X, Li Y, Pang P, Shu Z, Gong X. The Nomogram of MRI-based Radiomics with Complementary Visual Features by Machine Learning Improves Stratification of Glioblastoma Patients: A Multicenter Study. J Magn Reson Imaging 2021; 54:571-583. [PMID: 33559302 DOI: 10.1002/jmri.27536] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 01/15/2021] [Accepted: 01/16/2021] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Glioblastomas (GBMs) represent both the most common and the most highly malignant primary brain tumors. The subjective visual imaging features from MRI make it challenging to predict the overall survival (OS) of GBM. Radiomics can quantify image features objectively as an emerging technique. A pragmatic and objective method in the clinic to assess OS is strongly in need. PURPOSE To construct a radiomics nomogram to stratify GBM patients into long- vs. short-term survival. STUDY TYPE Retrospective. POPULATION One-hundred and fifty-eight GBM patients from Brain Tumor Segmentation Challenge 2018 (BRATS2018) were for model construction and 32 GBM patients from the local hospital for external validation. FIELD STRENGTH/SEQUENCE 1.5 T and 3.0 T MRI Scanners, T1 WI, T2 WI, T2 FLAIR, and contrast-enhanced T1 WI sequences ASSESSMENT: All patients were divided into long-term or short-term based on a survival of greater or fewer than 12 months. All BRATS2018 subjects were divided into training and test sets, and images were assessed for ependymal and pia mater involvement (EPI) and multifocality by three experienced neuroradiologists. All tumor tissues from multiparametric MRI were fully automatically segmented into three subregions to calculate the radiomic features. Based on the training set, the most powerful radiomic features were selected to constitute radiomic signature. STATISTICAL TESTS Receiver operating characteristic (ROC) curve, sensitivity, specificity, and the Hosmer-Lemeshow test. RESULTS The nomogram had a survival prediction accuracy of 0.878 and 0.875, a specificity of 0.875 and 0.583, and a sensitivity of 0.704 and 0.833, respectively, in the training and test set. The ROC curve showed the accuracy of the nomogram, radiomic signature, age, and EPI for external validation set were 0.858, 0.826, 0.664, and 0.66 in the validate set, respectively. DATA CONCLUSION Radiomics nomogram integrated with radiomic signature, EPI, and age was found to be robust for the stratification of GBM patients into long- vs. short-term survival. LEVEL OF EVIDENCE 3 TECHNICAL EFFICACY STAGE: 2.
Collapse
Affiliation(s)
- Yuyun Xu
- Department of Radiology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Xiaodong He
- Department of Radiology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Yumei Li
- Department of Radiology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital of Hangzhou Medical College, Hangzhou, China
| | | | - Zhenyu Shu
- Department of Radiology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Xiangyang Gong
- Department of Radiology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital of Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
13
|
Weninger L, Haarburger C, Merhof D. Robustness of Radiomics for Survival Prediction of Brain Tumor Patients Depending on Resection Status. Front Comput Neurosci 2019; 13:73. [PMID: 31780915 PMCID: PMC6857096 DOI: 10.3389/fncom.2019.00073] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 10/09/2019] [Indexed: 12/30/2022] Open
Abstract
Prediction of overall survival based on multimodal MRI of brain tumor patients is a difficult problem. Although survival also depends on factors that cannot be assessed via preoperative MRI such as surgical outcome, encouraging results for MRI-based survival analysis have been published for different datasets. We assess if and how established radiomic approaches as well as novel methods can predict overall survival of brain tumor patients on the BraTS challenge dataset. This dataset consists of multimodal preoperative images of 211 glioblastoma patients from several institutions with reported resection status and known survival. In the official challenge setting, only patients with a reported gross total resection (GTR) are taken into account. We therefore evaluated previously published methods as well as different machine learning approaches on the BraTS dataset. For different types of resection status, these approaches are compared to a baseline, a linear regression on patient age only. This naive approach won the 3rd place out of 26 participants in the BraTS survival prediction challenge 2018. Previously published radiomic signatures show significant correlations and predictiveness to patient survival for patients with a reported subtotal resection. However, for patients with reported GTR, none of the evaluated approaches was able to outperform the age-only baseline in a cross-validation setting, explaining the poor performance of approaches based on radiomics in the BraTS challenge 2018.
Collapse
Affiliation(s)
- Leon Weninger
- Imaging and Computer Vision, RWTH Aachen University, Aachen, Germany
| | | | | |
Collapse
|
14
|
Supratentorial high-grade astrocytoma with leptomeningeal spread to the fourth ventricle: a lethal dissemination with dismal prognosis. J Neurooncol 2019; 142:253-261. [PMID: 30604394 DOI: 10.1007/s11060-018-03086-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Accepted: 12/26/2018] [Indexed: 12/12/2022]
Abstract
PURPOSE Leptomeningeal spread to the fourth ventricle (LSFV) from supratentorial high-grade astrocytoma (HGA) is rarely investigated. The incidence and prognostic merit of LSFV were analyzed in this study. METHODS A consecutive cohort of 175 patients with pathologically diagnosed HGA according to the 2016 WHO classification of brain tumors was enrolled. LSFV was defined as radiological occupation in the fourth ventricle at the moment of initial progression. Clinical, radiological, and pathological data were analyzed to explore the difference between HGA patients with and without LSFV. RESULTS There were 18 of 175 (10.3%) HGAs confirmed with LSFV. The difference of survival rate between patients with LSFV or not was significant in both overall survival (OS) (14.5 vs. 24 months, P = 0.0007) and post progression survival (PPS) (6.0 vs. 11.5 months, P = 0.0004), while no significant difference was observed in time to progression (TTP) (8.5 months vs. 9.5 months P = 0.6795). In the Cox multivariate analysis, LSFV was confirmed as an independent prognostic risk factor for OS (HR 2.06, P = 0.010). LSFV was correlated with younger age (P = 0.044), ventricle infringement of primary tumor (P < 0.001) and higher Ki-67 index (P = 0.013) in further analysis, and the latter two have been validated in the Logistic regression analysis (OR 18.16, P = 0.006; OR 4.04, P = 0.012, respectively). CONCLUSION LSFV was indicative of end-stage for supratentorial HGA patients, which shortened patients' PPS and OS instead of TTP. It's never too cautious to alert this lethal event when tumor harbored ventricle infringement and higher Ki-67 index in routine clinical course.
Collapse
|
15
|
Vallatos A, Al-Mubarak HFI, Birch JL, Galllagher L, Mullin JM, Gilmour L, Holmes WM, Chalmers AJ. Quantitative histopathologic assessment of perfusion MRI as a marker of glioblastoma cell infiltration in and beyond the peritumoral edema region. J Magn Reson Imaging 2018; 50:529-540. [PMID: 30569620 DOI: 10.1002/jmri.26580] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 10/26/2018] [Accepted: 10/26/2018] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Conventional MRI fails to detect regions of glioblastoma cell infiltration beyond the contrast-enhanced T1 solid tumor region, with infiltrating tumor cells often migrating along host blood vessels. PURPOSE To quantitatively and qualitatively analyze the correlation between perfusion MRI signal and tumor cell density in order to assess whether local perfusion perturbation could provide a useful biomarker of glioblastoma cell infiltration. STUDY TYPE Animal model. SUBJECTS Mice bearing orthotopic glioblastoma xenografts generated from a patient-derived glioblastoma cell line. FIELD STRENGTH/SEQUENCES 7T perfusion images acquired using a high signal-to-noise ratio (SNR) multiple boli arterial spin labeling sequence were compared with conventional MRI (T1 /T2 weighted, contrast-enhanced T1 , diffusion-weighted, and apparent diffusion coefficient). ASSESSMENT Immunohistochemistry sections were stained for human leukocyte antigen (probing human-derived tumor cells). To achieve quantitative MRI-tissue comparison, multiple histological slices cut in the MRI plane were stacked to produce tumor cell density maps acting as a "ground truth." STATISTICAL TESTS Sensitivity, specificity, accuracy, and Dice similarity indices were calculated and a two-tailed, paired t-test used for statistical analysis. RESULTS High comparison test results (Dice 0.62-0.72, Accuracy 0.86-0.88, Sensitivity 0.51-0.7, and Specificity 0.92-0.97) indicate a good segmentation for all imaging modalities and highlight the quality of the MRI tissue assessment protocol. Perfusion imaging exhibits higher sensitivity (0.7) than conventional MRI (0.51-0.61). MRI/histology voxel-to-voxel comparison revealed a negative correlation between tumor cell infiltration and perfusion at the tumor margins (P = 0.0004). DATA CONCLUSION These results demonstrate the ability of perfusion imaging to probe regions of low tumor cell infiltration while confirming the sensitivity limitations of conventional imaging modalities. The quantitative relationship between tumor cell density and perfusion identified in and beyond the edematous T2 hyperintensity region surrounding macroscopic tumor could be used to detect marginal tumor cell infiltration with greater accuracy. LEVEL OF EVIDENCE 1 Technical stage: 2 J. Magn. Reson. Imaging 2019;50:529-540.
Collapse
Affiliation(s)
- A Vallatos
- Glasgow Experimental MRI Centre, Institute of Neuroscience and Psychology, University of Glasgow, UK.,Centre for Clinical Brain Sciences, University of Edinburgh, UK
| | - H F I Al-Mubarak
- Glasgow Experimental MRI Centre, Institute of Neuroscience and Psychology, University of Glasgow, UK.,University of Misan, Iraq
| | - J L Birch
- Wolfson Wohl Translational Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, UK
| | - L Galllagher
- Glasgow Experimental MRI Centre, Institute of Neuroscience and Psychology, University of Glasgow, UK
| | - J M Mullin
- Glasgow Experimental MRI Centre, Institute of Neuroscience and Psychology, University of Glasgow, UK
| | - L Gilmour
- Wolfson Wohl Translational Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, UK
| | - W M Holmes
- Glasgow Experimental MRI Centre, Institute of Neuroscience and Psychology, University of Glasgow, UK
| | - A J Chalmers
- Wolfson Wohl Translational Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, UK
| |
Collapse
|
16
|
Calluaud G, Terrier LM, Mathon B, Destrieux C, Velut S, François P, Zemmoura I, Amelot A. Peritumoral Edema/Tumor Volume Ratio: A Strong Survival Predictor for Posterior Fossa Metastases. Neurosurgery 2018; 85:117-125. [DOI: 10.1093/neuros/nyy222] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 06/04/2018] [Indexed: 12/29/2022] Open
Affiliation(s)
| | - Louis-Marie Terrier
- CHRU de Tours, Department of Neurosurgery, Tours, France
- Université François-Rabelais de Tours, Inserm, iBrain, UMR U1253, Tours, France
| | - Bertrand Mathon
- CHU La Pitié-Salpétrière, Department of Neurosurgery, Paris, France
| | - Christophe Destrieux
- CHRU de Tours, Department of Neurosurgery, Tours, France
- Université François-Rabelais de Tours, Inserm, iBrain, UMR U1253, Tours, France
| | - Stéphane Velut
- CHRU de Tours, Department of Neurosurgery, Tours, France
- Université François-Rabelais de Tours, Inserm, iBrain, UMR U1253, Tours, France
| | | | - Ilyess Zemmoura
- CHRU de Tours, Department of Neurosurgery, Tours, France
- Université François-Rabelais de Tours, Inserm, iBrain, UMR U1253, Tours, France
| | - Aymeric Amelot
- CHRU de Tours, Department of Neurosurgery, Tours, France
- CHU La Pitié-Salpétrière, Department of Neurosurgery, Paris, France
| |
Collapse
|
17
|
Leu S, Boulay JL, Thommen S, Bucher HC, Stippich C, Mariani L, Bink A. Preoperative Two-Dimensional Size of Glioblastoma is Associated with Patient Survival. World Neurosurg 2018; 115:e448-e463. [PMID: 29678715 DOI: 10.1016/j.wneu.2018.04.067] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 04/09/2018] [Accepted: 04/10/2018] [Indexed: 10/17/2022]
Abstract
BACKGROUND Although tumor size affects survival of patients with lower-grade glioma, a prognostic effect on patients with glioblastoma remains to be established. METHODS We performed a retrospective analysis of 61 patients using volumetric data of tumor compartments of 61 patients obtained by preoperative magnetic resonance images using the visual ABC/2 method. Preoperative enhancing, nonenhancing, necrosis, and edema volume, the preoperative tumor area (TA) as a product of the 2 largest tumor diameters perpendicular to each other on axial T1-weighted postcontrast images, as well as postoperative enhancing residual volumes, were measured. Multivariable Cox proportional hazard models were used to associate these parameters with overall survival, adjusting for potential confounders. RESULTS The median preoperative enhancing tumor volume was 18.2 mL (interquartile range, 8.2-41.7 mL); the median remnant tumor volume was 1.3% (interquartile range, 0.0%-42.9%). During follow-up, 59 patients (92%) died; median survival time and median follow-up time were both 404 days. We found a statistically significant multiplicative effect of TA on survival: the hazard ratio (HR) was increased by 1.096 per unit increase of 200 mm2 (95% confidence interval [CI], 1.027-1.170; P < 0.01). The effect of remnant tumor on HR increased multiplicatively by 1.013 (95% CI, 1.001-1.026; P = 0.04) per unit increase of 1 log (day) and 1% in tumor remnant. HR associated with age at surgery increased by 1.503 per 5 years of age (95% CI, 1.243-1.817; P < 0.01). CONCLUSIONS Preoperative TA proved to be the only glioblastoma size parameter that affects patient survival.
Collapse
Affiliation(s)
- Severina Leu
- Department of Neurosurgery, University Hospital Basel, University of Basel, Basel, Switzerland; Brain Tumor Biology Laboratory, Department of Neurosurgery, University Hospital Basel, University of Basel, Basel, Switzerland.
| | - Jean-Louis Boulay
- Brain Tumor Biology Laboratory, Department of Neurosurgery, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Sarah Thommen
- Basel Institute for Clinical Epidemiology and Biostatistics, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Heiner C Bucher
- Basel Institute for Clinical Epidemiology and Biostatistics, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Christoph Stippich
- Division of Neuroradiology, Department of Radiology, University Hospital Basel, University of Basel, Basel, Switzerland; Clinic for Neuroradiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Luigi Mariani
- Department of Neurosurgery, University Hospital Basel, University of Basel, Basel, Switzerland; Brain Tumor Biology Laboratory, Department of Neurosurgery, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Andrea Bink
- Division of Neuroradiology, Department of Radiology, University Hospital Basel, University of Basel, Basel, Switzerland; Clinic for Neuroradiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
18
|
Chaddad A, Daniel P, Desrosiers C, Toews M, Abdulkarim B. Novel Radiomic Features Based on Joint Intensity Matrices for Predicting Glioblastoma Patient Survival Time. IEEE J Biomed Health Inform 2018; 23:795-804. [PMID: 29993848 DOI: 10.1109/jbhi.2018.2825027] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
This paper presents a novel set of image texture features generalizing standard grey-level co-occurrence matrices (GLCM) to multimodal image data through joint intensity matrices (JIMs). These are used to predict the survival of glioblastoma multiforme (GBM) patients from multimodal MRI data. The scans of 73 GBM patients from the Cancer Imaging Archive are used in our study. Necrosis, active tumor, and edema/invasion subregions of GBM phenotypes are segmented using the coregistration of contrast-enhanced T1-weighted (CE-T1) images and its corresponding fluid-attenuated inversion recovery (FLAIR) images. Texture features are then computed from the JIM of these GBM subregions and a random forest model is employed to classify patients into short or long survival groups. Our survival analysis identified JIM features in necrotic (e.g., entropy and inverse-variance) and edema (e.g., entropy and contrast) subregions that are moderately correlated with survival time (i.e., Spearman rank correlation of 0.35). Moreover, nine features were found to be associated with GBM survival with a Hazard-ratio range of 0.38-2.1 and a significance level of p < 0.05 following Holm-Bonferroni correction. These features also led to the highest accuracy in a univariate analysis for predicting the survival group of patients, with AUC values in the range of 68-70%. Considering multiple features for this task, JIM features led to significantly higher AUC values than those based on standard GLCMs and gene expression. Furthermore, an AUC of 77.56% with p = 0.003 was achieved when combining JIM, GLCM, and gene expression features into a single radiogenomic signature. In summary, our study demonstrated the usefulness of modeling the joint intensity characteristics of CE-T1 and FLAIR images for predicting the prognosis of patients with GBM.
Collapse
|
19
|
Hou Z, Cai X, Li H, Zeng C, Wang J, Gao Z, Zhang M, Dou W, Zhang N, Zhang L, Xie J. Quantitative Assessment of Invasion of High-Grade Gliomas Using Diffusion Tensor Magnetic Resonance Imaging. World Neurosurg 2018; 113:e561-e567. [PMID: 29482009 DOI: 10.1016/j.wneu.2018.02.095] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 02/14/2018] [Accepted: 02/15/2018] [Indexed: 02/07/2023]
Abstract
OBJECTIVE To determine heterogeneity of high-grade glioma (HGG) and its surrounding area and explore quantitative analysis of invasion of HGG using diffusion tensor imaging. METHODS This study included 14 patients with HGG and preoperative magnetic resonance imaging and diffusion tensor imaging examinations. Three regions of interest were placed. Apparent diffusion coefficient (ADC) and fractional anisotropy (FA) values of these regions of interest were measured, and specimens from the 3 regions of interest were obtained under navigation guidance. Postoperative examinations of specimens were carried out. Correlations between ADC and FA values and tumor cell density were evaluated. RESULTS Median survival was 36.7 months. As distance from the tumor increased, the number of tumor cells significantly decreased. Regarding levels of matrix metalloproteinase-9 and Ki-67, only the differences between tumor and distances of 1 cm and 2 cm away from the tumor were statistically significant. For analysis of the relationship between tumor cell density and ADC and FA values, the discriminant formulas were as follows: G1 = -13.678 + 14984.791 (X) + 14443.847 (Y) (tumor cell density ≥10%); G2 = -11.649 + 14443.847 (X) + 33.285 (Y) (tumor cell density <10%). CONCLUSIONS We verified the heterogeneity of HGG and its surrounding area and found that patients with extensive resection may have longer survival. We also found a few formulas using FA and ADC values to predict tumor cell density.
Collapse
Affiliation(s)
- Zonggang Hou
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Xu Cai
- Department of Neurosurgery, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Huan Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Chun Zeng
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Jiangfei Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Zhixian Gao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Mingyu Zhang
- Department of Radiology, Beijing Neurosurgical Institute, Beijing, China
| | - Weibei Dou
- Department of Electronic Engineering, Tsinghua University, Beijing, China
| | - Ning Zhang
- Health Management and Education Institute, Capital Medical University, Beijing, China
| | - Liwei Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Jian Xie
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; China National Clinical Research Center for Neurological Diseases, Beijing, China.
| |
Collapse
|
20
|
Steed TC, Treiber JM, Brandel MG, Patel KS, Dale AM, Carter BS, Chen CC. Quantification of glioblastoma mass effect by lateral ventricle displacement. Sci Rep 2018; 8:2827. [PMID: 29434275 PMCID: PMC5809591 DOI: 10.1038/s41598-018-21147-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 01/26/2018] [Indexed: 11/08/2022] Open
Abstract
Mass effect has demonstrated prognostic significance for glioblastoma, but is poorly quantified. Here we define and characterize a novel neuroimaging parameter, lateral ventricle displacement (LVd), which quantifies mass effect in glioblastoma patients. LVd is defined as the magnitude of displacement from the center of mass of the lateral ventricle volume in glioblastoma patients relative to that a normal reference brain. Pre-operative MR images from 214 glioblastoma patients from The Cancer Imaging Archive (TCIA) were segmented using iterative probabilistic voxel labeling (IPVL). LVd, contrast enhancing volumes (CEV) and FLAIR hyper-intensity volumes (FHV) were determined. Associations with patient survival and tumor genomics were investigated using data from The Cancer Genome Atlas (TCGA). Glioblastoma patients had significantly higher LVd relative to patients without brain tumors. The variance of LVd was not explained by tumor volume, as defined by CEV or FLAIR. LVd was robustly associated with glioblastoma survival in Cox models which accounted for both age and Karnofsky's Performance Scale (KPS) (p = 0.006). Glioblastomas with higher LVd demonstrated increased expression of genes associated with tumor proliferation and decreased expression of genes associated with tumor invasion. Our results suggest LVd is a quantitative measure of glioblastoma mass effect and a prognostic imaging biomarker.
Collapse
Affiliation(s)
- Tyler C Steed
- Department of Neurosurgery, Emory University, Atlanta, GA, USA
| | - Jeffrey M Treiber
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX, USA
| | - Michael G Brandel
- Department of Neurosurgery, University of California San Diego, La Jolla, CA, USA
| | - Kunal S Patel
- Department of Neurosurgery, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
| | - Anders M Dale
- Multimodal Imaging Laboratory, University of California San Diego, La Jolla, CA, USA
- Department of Radiology, University of California San Diego, La Jolla, CA, USA
| | - Bob S Carter
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, USA
| | - Clark C Chen
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
21
|
Amelot A, Deroulers C, Badoual M, Polivka M, Adle-Biassette H, Houdart E, Carpentier AF, Froelich S, Mandonnet E. Surgical Decision Making From Image-Based Biophysical Modeling of Glioblastoma: Not Ready for Primetime. Neurosurgery 2018; 80:793-799. [PMID: 28387870 DOI: 10.1093/neuros/nyw186] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2016] [Accepted: 03/17/2017] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Biophysical modeling of glioma is gaining more interest for clinical practice. The most popular model describes aggressivity of tumor cells by two parameters: net proliferation rate (ρ) and propensity to migrate (D). The ratio ρ/D, which can be estimated from a single preoperative magnetic resonance imaging (MRI), characterizes tumor invasiveness profile (high ρ/D: nodular; low ρ/D: diffuse). A recent study reported, from a large series of glioblastoma multiforme (GBM) patients, that gross total resection (GTR) would improve survival only in patients with nodular tumors. OBJECTIVE To replicate these results, that is to verify that benefit of GTR would be only observed for nodular tumors. METHODS Between 2005 and 2012, we considered 234 GBM patients with pre- and postoperative MRI. Stereotactic biopsy (BST) was performed in 109 patients. Extent of resection was assessed on postoperative MRI and classified as GTR or partial resection (PR). Invasiveness ρ/D was estimated from the preoperative tumor volumes on T1-Gadolinium-enhanced and fluid-attenuated inversion recovery sequences. RESULTS We demonstrate that patients with diffuse GBM (low ρ/D), as well as more nodular (mid and high ρ/D) GBM, presented significant survival benefit from GTR over PR/BST ( P < .001). CONCLUSION Whatever the degree of tumor invasiveness, as estimated from MRI-driven biophysical modeling, GTR improves survival of GBM patients, compared to PR or BST. This conflicting result should motivate further studies.
Collapse
Affiliation(s)
- Aymeric Amelot
- Assistance Publique-Hôpitaux de Paris (AP-HP), Service de Neurochirurgie, Hôpital Lariboisière, Paris, France
| | | | | | - Marc Polivka
- Assistance Publique-Hôpitaux de Paris (AP-HP), Service d'Anatomopathologie, Hôpital Lariboisière, Paris, France
| | - Homa Adle-Biassette
- Assistance Publique-Hôpitaux de Paris (AP-HP), Service d'Anatomopathologie, Hôpital Lariboisière, Paris, France
| | - Emmanuel Houdart
- Assistance Publique-Hôpitaux de Paris (AP-HP), Service de Neuroradiologie, Hôpital Lariboisière, Paris, France
| | - Antoine F Carpentier
- Assistance Publique-Hôpitaux de Paris (AP-HP), Service de Neurologie, Hôpital Avicenne, Bobigny, France
| | - Sebastien Froelich
- Assistance Publique-Hôpitaux de Paris (AP-HP), Service de Neurochirurgie, Hôpital Lariboisière, Paris, France.,Université Paris 7 Diderot, Paris, France
| | - Emmanuel Mandonnet
- Assistance Publique-Hôpitaux de Paris (AP-HP), Service de Neurochirurgie, Hôpital Lariboisière, Paris, France.,IMNC, UMR8165, Orsay, France.,Université Paris 7 Diderot, Paris, France
| |
Collapse
|
22
|
Beig N, Patel J, Prasanna P, Hill V, Gupta A, Correa R, Bera K, Singh S, Partovi S, Varadan V, Ahluwalia M, Madabhushi A, Tiwari P. Radiogenomic analysis of hypoxia pathway is predictive of overall survival in Glioblastoma. Sci Rep 2018; 8:7. [PMID: 29311558 PMCID: PMC5758516 DOI: 10.1038/s41598-017-18310-0] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 12/04/2017] [Indexed: 12/24/2022] Open
Abstract
Hypoxia, a characteristic trait of Glioblastoma (GBM), is known to cause resistance to chemo-radiation treatment and is linked with poor survival. There is hence an urgent need to non-invasively characterize tumor hypoxia to improve GBM management. We hypothesized that (a) radiomic texture descriptors can capture tumor heterogeneity manifested as a result of molecular variations in tumor hypoxia, on routine treatment naïve MRI, and (b) these imaging based texture surrogate markers of hypoxia can discriminate GBM patients as short-term (STS), mid-term (MTS), and long-term survivors (LTS). 115 studies (33 STS, 41 MTS, 41 LTS) with gadolinium-enhanced T1-weighted MRI (Gd-T1w) and T2-weighted (T2w) and FLAIR MRI protocols and the corresponding RNA sequences were obtained. After expert segmentation of necrotic, enhancing, and edematous/nonenhancing tumor regions for every study, 30 radiomic texture descriptors were extracted from every region across every MRI protocol. Using the expression profile of 21 hypoxia-associated genes, a hypoxia enrichment score (HES) was obtained for the training cohort of 85 cases. Mutual information score was used to identify a subset of radiomic features that were most informative of HES within 3-fold cross-validation to categorize studies as STS, MTS, and LTS. When validated on an additional cohort of 30 studies (11 STS, 9 MTS, 10 LTS), our results revealed that the most discriminative features of HES were also able to distinguish STS from LTS (p = 0.003).
Collapse
Affiliation(s)
- Niha Beig
- Case Western Reserve University, Department of Biomedical Engineering, Cleveland, 44106, USA.
| | - Jay Patel
- Case Western Reserve University, Department of Biomedical Engineering, Cleveland, 44106, USA
| | - Prateek Prasanna
- Case Western Reserve University, Department of Biomedical Engineering, Cleveland, 44106, USA
| | - Virginia Hill
- Department of Neuroradiology, Imaging Institute, Cleveland Clinic, Cleveland, 44106, USA
| | - Amit Gupta
- University Hospitals of Cleveland, Department of Radiology, Cleveland, 44106, USA
| | - Ramon Correa
- Case Western Reserve University, Department of Biomedical Engineering, Cleveland, 44106, USA
| | - Kaustav Bera
- Case Western Reserve University, Department of Biomedical Engineering, Cleveland, 44106, USA
| | - Salendra Singh
- Case Western Reserve University, School of Medicine, Cleveland, 44106, USA
| | - Sasan Partovi
- University Hospitals of Cleveland, Department of Radiology, Cleveland, 44106, USA
| | - Vinay Varadan
- Case Western Reserve University, School of Medicine, Cleveland, 44106, USA
| | - Manmeet Ahluwalia
- Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, 44106, USA
| | - Anant Madabhushi
- Case Western Reserve University, Department of Biomedical Engineering, Cleveland, 44106, USA
| | - Pallavi Tiwari
- Case Western Reserve University, Department of Biomedical Engineering, Cleveland, 44106, USA.
| |
Collapse
|
23
|
Quantitative radiomic profiling of glioblastoma represents transcriptomic expression. Oncotarget 2018; 9:6336-6345. [PMID: 29464076 PMCID: PMC5814216 DOI: 10.18632/oncotarget.23975] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 09/06/2017] [Indexed: 01/29/2023] Open
Abstract
Quantitative imaging biomarkers have increasingly emerged in the field of research utilizing available imaging modalities. We aimed to identify good surrogate radiomic features that can represent genetic changes of tumors, thereby establishing noninvasive means for predicting treatment outcome. From May 2012 to June 2014, we retrospectively identified 65 patients with treatment-naïve glioblastoma with available clinical information from the Samsung Medical Center data registry. Preoperative MR imaging data were obtained for all 65 patients with primary glioblastoma. A total of 82 imaging features including first-order statistics, volume, and size features, were semi-automatically extracted from structural and physiologic images such as apparent diffusion coefficient and perfusion images. Using commercially available software, NordicICE, we performed quantitative imaging analysis and collected the dataset composed of radiophenotypic parameters. Unsupervised clustering methods revealed that the radiophenotypic dataset was composed of three clusters. Each cluster represented a distinct molecular classification of glioblastoma; classical type, proneural and neural types, and mesenchymal type. These clusters also reflected differential clinical outcomes. We found that extracted imaging signatures does not represent copy number variation and somatic mutation. Quantitative radiomic features provide a potential evidence to predict molecular phenotype and treatment outcome. Radiomic profiles represents transcriptomic phenotypes more well.
Collapse
|
24
|
Prasanna P, Patel J, Partovi S, Madabhushi A, Tiwari P. Radiomic features from the peritumoral brain parenchyma on treatment-naïve multi-parametric MR imaging predict long versus short-term survival in glioblastoma multiforme: Preliminary findings. Eur Radiol 2017; 27:4188-4197. [PMID: 27778090 PMCID: PMC5403632 DOI: 10.1007/s00330-016-4637-3] [Citation(s) in RCA: 195] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 09/28/2016] [Accepted: 10/05/2016] [Indexed: 10/20/2022]
Abstract
OBJECTIVE Despite 90 % of glioblastoma (GBM) recurrences occurring in the peritumoral brain zone (PBZ), its contribution in patient survival is poorly understood. The current study leverages computerized texture (i.e. radiomic) analysis to evaluate the efficacy of PBZ features from pre-operative MRI in predicting long- (>18 months) versus short-term (<7 months) survival in GBM. METHODS Sixty-five patient examinations (29 short-term, 36 long-term) with gadolinium-contrast T1w, FLAIR and T2w sequences from the Cancer Imaging Archive were employed. An expert manually segmented each study as: enhancing lesion, PBZ and tumour necrosis. 402 radiomic features (capturing co-occurrence, grey-level dependence and directional gradients) were obtained for each region. Evaluation was performed using threefold cross-validation, such that a subset of studies was used to select the most predictive features, and the remaining subset was used to evaluate their efficacy in predicting survival. RESULTS A subset of ten radiomic 'peritumoral' MRI features, suggestive of intensity heterogeneity and textural patterns, was found to be predictive of survival (p = 1.47 × 10-5) as compared to features from enhancing tumour, necrotic regions and known clinical factors. CONCLUSION Our preliminary analysis suggests that radiomic features from the PBZ on routine pre-operative MRI may be predictive of long- versus short-term survival in GBM. KEY POINTS • Radiomic features from peritumoral regions can capture glioblastoma heterogeneity to predict outcome. • Peritumoral radiomics along with clinical factors are highly predictive of glioblastoma outcome. • Identifying prognostic markers can assist in making personalized therapy decisions in glioblastoma.
Collapse
Affiliation(s)
- Prateek Prasanna
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH, 44106, USA
| | - Jay Patel
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH, 44106, USA
| | - Sasan Partovi
- Case Western Reserve School of Medicine, University Hospitals Case Medical Center, 11100 Euclid Ave, Cleveland, OH, 44106, USA
| | - Anant Madabhushi
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH, 44106, USA
| | - Pallavi Tiwari
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH, 44106, USA.
| |
Collapse
|
25
|
Garrett MD, Yanagihara TK, Yeh R, McKhann GM, Sisti MB, Bruce JN, Sheth SA, Sonabend AM, Wang TJC. Monitoring Radiation Treatment Effects in Glioblastoma: FLAIR Volume as Significant Predictor of Survival. Tomography 2017; 3:131-137. [PMID: 30042977 PMCID: PMC6024439 DOI: 10.18383/j.tom.2017.00009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Glioblastoma is the most common adult central nervous system malignancy and carries a poor prognosis. Disease progression and recurrence after chemoradiotherapy are assessed via serial magnetic resonance imaging sequences. T2-weighted fluid-attenuated inversion recovery (FLAIR) signal is presumed to represent edema containing microscopic cancer infiltration. Here we assessed the prognostic impact of computerized volumetry of FLAIR signal in the peri-treatment setting for glioblastoma. We analyzed pre- and posttreatment FLAIR sequences of 40 patients treated at the Columbia University Medical Center between 2011 and 2014, excluding those without high-quality FLAIR imaging within 2 weeks before treatment and 60 to 180 days afterward. We manually contoured regions of FLAIR hyperintensity as per Radiation Therapy Oncology Group guidelines and calculated the volumes of nonenhancing tumor burden. At the time of this study, all but 1 patient had died. Pre- and posttreatment FLAIR volumes were assessed for correlation to overall and progression-free survival. Larger post-treatment FLAIR volumes from sequences taken between 60 and 180 days after conclusion of chemoradiotherapy were negatively correlated with overall survival (P = .048 on Pearson's correlation and P = .017 and P = .043 on univariable and multivariable Cox regression analyses, respectively) and progression-free survival (P = .002 on Pearson's correlation and P = < .001 and P = < .001 on univariable and multivariable Cox regression analyses). This study suggests that higher FLAIR volumes in the 2- to 6-month posttreatment window are associated with worsened survival.
Collapse
Affiliation(s)
| | | | | | - Guy M. McKhann
- Neurological Surgery, Columbia University Medical Center, New York, NY; and,Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY
| | - Michael B. Sisti
- Neurological Surgery, Columbia University Medical Center, New York, NY; and,Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY
| | - Jeffrey N. Bruce
- Neurological Surgery, Columbia University Medical Center, New York, NY; and,Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY
| | - Sameer A. Sheth
- Neurological Surgery, Columbia University Medical Center, New York, NY; and,Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY
| | - Adam M. Sonabend
- Neurological Surgery, Columbia University Medical Center, New York, NY; and,Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY
| | - Tony J. C. Wang
- Radiation Oncology;,Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY
| |
Collapse
|
26
|
Abstract
The imaging of treated gliomas is complicated by a variety of treatment related effects, which can falsely simulate disease improvement or progression. Distinguishing between disease progression and treatment effects is difficult with standard MR imaging pulse sequences and added specificity can be gained by the addition of advanced imaging techniques.
Collapse
Affiliation(s)
- Mark F Dalesandro
- Department of Radiology, Harborview Medical Center, University of Washington, Box 357115, 1959 Northeast Pacific Street, NW011, Seattle, WA 98195-7115, USA
| | - Jalal B Andre
- Department of Radiology, Harborview Medical Center, University of Washington, Box 357115, 1959 Northeast Pacific Street, NW011, Seattle, WA 98195-7115, USA.
| |
Collapse
|
27
|
Jiang H, Cui Y, Liu X, Ren X, Lin S. Patient-Specific Resection Strategy of Glioblastoma Multiforme: Choice Based on a Preoperative Scoring Scale. Ann Surg Oncol 2017; 24:2006-2014. [PMID: 28321691 DOI: 10.1245/s10434-017-5843-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Indexed: 11/18/2022]
Abstract
BACKGROUND The real association between extent of resection and outcome in patients with glioblastoma multiforme (GBM) remains unclear. OBJECTIVE The goal of this study was to disclose the effect of gross total resection on survival and establish a scale used for surgical decision making. METHODS A retrospective review was undertaken of 416 patients who received operation for GBM from 2008 to 2015 in Beijing Tiantan Hospital. To reduce bias in patient selection, propensity score analysis was conducted and 99 pairs of matched GBMs were generated. Survival between different groups was compared using the Kaplan-Meier method, and independent predictors of survival were identified using the Cox proportional hazards model. RESULTS Overall, the survival of patients undergoing GTR was significantly longer than those not undergoing GTR (12.0 vs. 9.0 months [p < 0.001] for progression-free survival [PFS], and 20.5 versus 16.0 months [p < 0.001] for overall survival [OS]). In the propensity model, the survival benefit of GTR remained significant, which has been further validated in the multivariate analysis (hazard ratio [HR] 0.613, 95% confidence interval [CI] 0.454-0.827 [p = 0.001] for PFS, and HR 0.475, 95% CI 0.343-0.659 [p < 0.001] for OS). Using a scoring scale based on age, epilepsy, location, tumor size, and Karnofsky performance score, patients were stratified into low-, moderate-, and high-risk cohorts. The survival benefit of GTR could be observed in the low- and moderate-risk cohorts but not the high-risk cohort. CONCLUSION GTR was an independent predictor of increased survival for patients with GBM. The risk scoring scale quantified the clinical significance of operation and helped us to project more personalized surgical strategies for individual patients.
Collapse
Affiliation(s)
- Haihui Jiang
- Department of Neurosurgery, First Hospital of Tsinghua University, Beijing, China
| | - Yong Cui
- Department of Neurosurgery, Beijing Tiantan Hospital, China National Clinical Research Center for Neurological Diseases, Center of Brain Tumor, Beijing Institute for Brain Disorders and Beijing Key Laboratory of Brain Tumor, Capital Medical University, Beijing, China
| | - Xiang Liu
- Department of Imaging Sciences, University of Rochester Medical Center, Rochester, NY, USA
| | - Xiaohui Ren
- Department of Neurosurgery, Beijing Tiantan Hospital, China National Clinical Research Center for Neurological Diseases, Center of Brain Tumor, Beijing Institute for Brain Disorders and Beijing Key Laboratory of Brain Tumor, Capital Medical University, Beijing, China
| | - Song Lin
- Department of Neurosurgery, Beijing Tiantan Hospital, China National Clinical Research Center for Neurological Diseases, Center of Brain Tumor, Beijing Institute for Brain Disorders and Beijing Key Laboratory of Brain Tumor, Capital Medical University, Beijing, China.
| |
Collapse
|
28
|
|
29
|
Fyllingen EH, Stensjøen AL, Berntsen EM, Solheim O, Reinertsen I. Glioblastoma Segmentation: Comparison of Three Different Software Packages. PLoS One 2016; 11:e0164891. [PMID: 27780224 PMCID: PMC5079567 DOI: 10.1371/journal.pone.0164891] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 10/03/2016] [Indexed: 11/18/2022] Open
Abstract
To facilitate a more widespread use of volumetric tumor segmentation in clinical studies, there is an urgent need for reliable, user-friendly segmentation software. The aim of this study was therefore to compare three different software packages for semi-automatic brain tumor segmentation of glioblastoma; namely BrainVoyagerTM QX, ITK-Snap and 3D Slicer, and to make data available for future reference. Pre-operative, contrast enhanced T1-weighted 1.5 or 3 Tesla Magnetic Resonance Imaging (MRI) scans were obtained in 20 consecutive patients who underwent surgery for glioblastoma. MRI scans were segmented twice in each software package by two investigators. Intra-rater, inter-rater and between-software agreement was compared by using differences of means with 95% limits of agreement (LoA), Dice’s similarity coefficients (DSC) and Hausdorff distance (HD). Time expenditure of segmentations was measured using a stopwatch. Eighteen tumors were included in the analyses. Inter-rater agreement was highest for BrainVoyager with difference of means of 0.19 mL and 95% LoA from -2.42 mL to 2.81 mL. Between-software agreement and 95% LoA were very similar for the different software packages. Intra-rater, inter-rater and between-software DSC were ≥ 0.93 in all analyses. Time expenditure was approximately 41 min per segmentation in BrainVoyager, and 18 min per segmentation in both 3D Slicer and ITK-Snap. Our main findings were that there is a high agreement within and between the software packages in terms of small intra-rater, inter-rater and between-software differences of means and high Dice’s similarity coefficients. Time expenditure was highest for BrainVoyager, but all software packages were relatively time-consuming, which may limit usability in an everyday clinical setting.
Collapse
Affiliation(s)
- Even Hovig Fyllingen
- Department of Neurosurgery, St. Olav’s University Hospital, Trondheim, Norway
- * E-mail: (EHF); (ALS)
| | - Anne Line Stensjøen
- Department of Circulation and Medical Imaging, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- * E-mail: (EHF); (ALS)
| | - Erik Magnus Berntsen
- Department of Circulation and Medical Imaging, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Radiology and Nuclear Medicine, St. Olav’s University Hospital, Trondheim, Norway
| | - Ole Solheim
- Department of Neurosurgery, St. Olav’s University Hospital, Trondheim, Norway
- Department of Neuroscience, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Ingerid Reinertsen
- Department of Neurosurgery, St. Olav’s University Hospital, Trondheim, Norway
- SINTEF, Technology and Society, Dept. Medical technology, Trondheim, Norway
| |
Collapse
|
30
|
Dunn WD, Aerts HJ, Cooper LA, Holder CA, Hwang SN, Jaffe CC, Brat DJ, Jain R, Flanders AE, Zinn PO, Colen RR, Gutman DA. Assessing the Effects of Software Platforms on Volumetric Segmentation of Glioblastoma. JOURNAL OF NEUROIMAGING IN PSYCHIATRY & NEUROLOGY 2016; 1:64-72. [PMID: 29600296 PMCID: PMC5870135 DOI: 10.17756/jnpn.2016-008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Radiological assessments of biologically relevant regions in glioblastoma have been associated with genotypic characteristics, implying a potential role in personalized medicine. Here, we assess the reproducibility and association with survival of two volumetric segmentation platforms and explore how methodology could impact subsequent interpretation and analysis. METHODS Post-contrast T1- and T2-weighted FLAIR MR images of 67 TCGA patients were segmented into five distinct compartments (necrosis, contrast-enhancement, FLAIR, post contrast abnormal, and total abnormal tumor volumes) by two quantitative image segmentation platforms - 3D Slicer and a method based on Velocity AI and FSL. We investigated the internal consistency of each platform by correlation statistics, association with survival, and concordance with consensus neuroradiologist ratings using ordinal logistic regression. RESULTS We found high correlations between the two platforms for FLAIR, post contrast abnormal, and total abnormal tumor volumes (spearman's r(67) = 0.952, 0.959, and 0.969 respectively). Only modest agreement was observed for necrosis and contrast-enhancement volumes (r(67) = 0.693 and 0.773 respectively), likely arising from differences in manual and automated segmentation methods of these regions by 3D Slicer and Velocity AI/FSL, respectively. Survival analysis based on AUC revealed significant predictive power of both platforms for the following volumes: contrast-enhancement, post contrast abnormal, and total abnormal tumor volumes. Finally, ordinal logistic regression demonstrated correspondence to manual ratings for several features. CONCLUSION Tumor volume measurements from both volumetric platforms produced highly concordant and reproducible estimates across platforms for general features. As automated or semi-automated volumetric measurements replace manual linear or area measurements, it will become increasingly important to keep in mind that measurement differences between segmentation platforms for more detailed features could influence downstream survival or radio genomic analyses.
Collapse
Affiliation(s)
- William D. Dunn
- Departments of Biomedical Informatics and Neurology, Emory
University School of Medicine, Atlanta, GA, USA
| | - Hugo J.W.L. Aerts
- Departments of Radiation Oncology and Radiology, Dana-Farber Cancer
Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA,
USA
- Department of Biostatistics & Computational Biology, Dana-Farber
Cancer Institute, Boston, MA, USA
| | - Lee A. Cooper
- Departments of Biomedical Informatics and Neurology, Emory
University School of Medicine, Atlanta, GA, USA
- Department Winship Cancer Institute, Emory University, Atlanta, GA,
USA
- Department Biomedical Engineering, Georgia Institute of
Technology/Emory University, Atlanta, GA, USA
| | - Chad A. Holder
- Department of Radiology and Imaging Sciences, Emory University
School of Medicine, Atlanta, GA, USA
| | - Scott N. Hwang
- Department of Diagnostic Imaging Department, St. Jude
Children’s Research Hospital, Memphis, TN, USA
| | - Carle C. Jaffe
- Department of Radiology, Boston University School of Medicine,
Boston, MA, USA
| | - Daniel J. Brat
- Department of Pathology and Laboratory Medicine, Emory University
School of Medicine, Atlanta, GA, USA
| | - Rajan Jain
- Departments of Radiology and Neurosurgery, NYU School of Medicine,
New York, NY, USA
| | - Adam E. Flanders
- Department of Neuroradiology, Thomas Jefferson University
Hospitals, Philadelphia, PA, USA
| | - Pascal O. Zinn
- Department of Neurosurgery, The University of Texas MD Anderson
Cancer Center, Houston, TX, USA
| | - Rivka R. Colen
- Department of Diagnostic Radiology, The University of Texas MD
Anderson Cancer Center, Houston, TX, USA
| | - David A. Gutman
- Departments of Biomedical Informatics and Neurology, Emory
University School of Medicine, Atlanta, GA, USA
- Department Winship Cancer Institute, Emory University, Atlanta, GA,
USA
| |
Collapse
|
31
|
Integrative analysis of diffusion-weighted MRI and genomic data to inform treatment of glioblastoma. J Neurooncol 2016; 129:289-300. [PMID: 27393347 DOI: 10.1007/s11060-016-2174-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 06/04/2016] [Indexed: 12/15/2022]
Abstract
Gene expression profiling from glioblastoma (GBM) patients enables characterization of cancer into subtypes that can be predictive of response to therapy. An integrative analysis of imaging and gene expression data can potentially be used to obtain novel biomarkers that are closely associated with the genetic subtype and gene signatures and thus provide a noninvasive approach to stratify GBM patients. In this retrospective study, we analyzed the expression of 12,042 genes for 558 patients from The Cancer Genome Atlas (TCGA). Among these patients, 50 patients had magnetic resonance imaging (MRI) studies including diffusion weighted (DW) MRI in The Cancer Imaging Archive (TCIA). We identified the contrast enhancing region of the tumors using the pre- and post-contrast T1-weighted MRI images and computed the apparent diffusion coefficient (ADC) histograms from the DW-MRI images. Using the gene expression data, we classified patients into four molecular subtypes, determined the number and composition of genes modules using the gap statistic, and computed gene signature scores. We used logistic regression to find significant predictors of GBM subtypes. We compared the predictors for different subtypes using Mann-Whitney U tests. We assessed detection power using area under the receiver operating characteristic (ROC) analysis. We computed Spearman correlations to determine the associations between ADC and each of the gene signatures. We performed gene enrichment analysis using Ingenuity Pathway Analysis (IPA). We adjusted all p values using the Benjamini and Hochberg method. The mean ADC was a significant predictor for the neural subtype. Neural tumors had a significantly lower mean ADC compared to non-neural tumors ([Formula: see text]), with mean ADC of [Formula: see text] and [Formula: see text] for neural and non-neural tumors, respectively. Mean ADC showed an area under the ROC of 0.75 for detecting neural tumors. We found eight gene modules in the GBM cohort. The mean ADC was significantly correlated with the gene signature related with dendritic cell maturation ([Formula: see text], [Formula: see text]). Mean ADC could be used as a biomarker of a gene signature associated with dendritic cell maturation and to assist in identifying patients with neural GBMs, known to be resistant to aggressive standard of care.
Collapse
|
32
|
Pérez-Beteta J, Martínez-González A, Molina D, Amo-Salas M, Luque B, Arregui E, Calvo M, Borrás JM, López C, Claramonte M, Barcia JA, Iglesias L, Avecillas J, Albillo D, Navarro M, Villanueva JM, Paniagua JC, Martino J, Velásquez C, Asenjo B, Benavides M, Herruzo I, Delgado MDC, Del Valle A, Falkov A, Schucht P, Arana E, Pérez-Romasanta L, Pérez-García VM. Glioblastoma: does the pre-treatment geometry matter? A postcontrast T1 MRI-based study. Eur Radiol 2016; 27:1096-1104. [PMID: 27329522 DOI: 10.1007/s00330-016-4453-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 05/11/2016] [Accepted: 05/25/2016] [Indexed: 10/21/2022]
Abstract
BACKGROUND The potential of a tumour's volumetric measures obtained from pretreatment MRI sequences of glioblastoma (GBM) patients as predictors of clinical outcome has been controversial. Mathematical models of GBM growth have suggested a relation between a tumour's geometry and its aggressiveness. METHODS A multicenter retrospective clinical study was designed to study volumetric and geometrical measures on pretreatment postcontrast T1 MRIs of 117 GBM patients. Clinical variables were collected, tumours segmented, and measures computed including: contrast enhancing (CE), necrotic, and total volumes; maximal tumour diameter; equivalent spherical CE width and several geometric measures of the CE "rim". The significance of the measures was studied using proportional hazards analysis and Kaplan-Meier curves. RESULTS Kaplan-Meier and univariate Cox survival analysis showed that total volume [p = 0.034, Hazard ratio (HR) = 1.574], CE volume (p = 0.017, HR = 1.659), spherical rim width (p = 0.007, HR = 1.749), and geometric heterogeneity (p = 0.015, HR = 1.646) were significant parameters in terms of overall survival (OS). Multivariable Cox analysis for OS provided the later two parameters as age-adjusted predictors of OS (p = 0.043, HR = 1.536 and p = 0.032, HR = 1.570, respectively). CONCLUSION Patients with tumours having small geometric heterogeneity and/or spherical rim widths had significantly better prognosis. These novel imaging biomarkers have a strong individual and combined prognostic value for GBM patients. KEY POINTS • Three-dimensional segmentation on magnetic resonance images allows the study of geometric measures. • Patients with small width of contrast enhancing areas have better prognosis. • The irregularity of contrast enhancing areas predicts survival in glioblastoma patients.
Collapse
Affiliation(s)
- Julián Pérez-Beteta
- Laboratory of Mathematical Oncology, Edificio Politécnico, Instituto de Matemática Aplicada a la Ciencia y la Ingeniería, Universidad de Castilla-La Mancha, Avenida de Camilo José Cela, 3, 13071, Ciudad Real, Spain.
| | - Alicia Martínez-González
- Laboratory of Mathematical Oncology, Edificio Politécnico, Instituto de Matemática Aplicada a la Ciencia y la Ingeniería, Universidad de Castilla-La Mancha, Avenida de Camilo José Cela, 3, 13071, Ciudad Real, Spain
| | - David Molina
- Laboratory of Mathematical Oncology, Edificio Politécnico, Instituto de Matemática Aplicada a la Ciencia y la Ingeniería, Universidad de Castilla-La Mancha, Avenida de Camilo José Cela, 3, 13071, Ciudad Real, Spain
| | - Mariano Amo-Salas
- Laboratory of Mathematical Oncology, Edificio Politécnico, Instituto de Matemática Aplicada a la Ciencia y la Ingeniería, Universidad de Castilla-La Mancha, Avenida de Camilo José Cela, 3, 13071, Ciudad Real, Spain
| | - Belén Luque
- Laboratory of Mathematical Oncology, Edificio Politécnico, Instituto de Matemática Aplicada a la Ciencia y la Ingeniería, Universidad de Castilla-La Mancha, Avenida de Camilo José Cela, 3, 13071, Ciudad Real, Spain
| | - Elena Arregui
- Hospital General de Ciudad Real, c/ Obispo Rafael Torija, Ciudad Real, Spain
| | - Manuel Calvo
- Hospital General de Ciudad Real, c/ Obispo Rafael Torija, Ciudad Real, Spain
| | - José M Borrás
- Hospital General de Ciudad Real, c/ Obispo Rafael Torija, Ciudad Real, Spain
| | - Carlos López
- Hospital General de Ciudad Real, c/ Obispo Rafael Torija, Ciudad Real, Spain
| | - Marta Claramonte
- Hospital General de Ciudad Real, c/ Obispo Rafael Torija, Ciudad Real, Spain
| | | | | | | | - David Albillo
- Hospital Universitario de Salamanca, Salamanca, Spain
| | | | | | | | - Juan Martino
- Hospital Marqués de Valdecilla, Santander, Spain
| | | | | | | | | | | | - Ana Del Valle
- Facultad de Matemáticas, Universidad de Sevilla, Sevilla, Spain
| | | | | | | | | | - Víctor M Pérez-García
- Laboratory of Mathematical Oncology, Edificio Politécnico, Instituto de Matemática Aplicada a la Ciencia y la Ingeniería, Universidad de Castilla-La Mancha, Avenida de Camilo José Cela, 3, 13071, Ciudad Real, Spain
| |
Collapse
|
33
|
Chang K, Zhang B, Guo X, Zong M, Rahman R, Sanchez D, Winder N, Reardon DA, Zhao B, Wen PY, Huang RY. Multimodal imaging patterns predict survival in recurrent glioblastoma patients treated with bevacizumab. Neuro Oncol 2016; 18:1680-1687. [PMID: 27257279 DOI: 10.1093/neuonc/now086] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 03/30/2016] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Bevacizumab is a humanized antibody against vascular endothelial growth factor approved for treatment of recurrent glioblastoma. There is a need to discover imaging biomarkers that can aid in the selection of patients who will likely derive the most survival benefit from bevacizumab. METHODS The aim of the study was to examine if pre- and posttherapy multimodal MRI features could predict progression-free survival and overall survival (OS) for patients with recurrent glioblastoma treated with bevacizumab. The patient population included 84 patients in a training cohort and 42 patients in a testing cohort, separated based on pretherapy imaging date. Tumor volumes of interest were segmented from contrast-enhanced T1-weighted and fluid attenuated inversion recovery images and were used to derive volumetric, shape, texture, parametric, and histogram features. A total of 2293 pretherapy and 9811 posttherapy features were used to generate the model. RESULTS Using standard radiographic assessment criteria, the hazard ratio for predicting OS was 3.38 (P < .001). The hazard ratios for pre- and posttherapy features predicting OS were 5.10 (P < .001) and 3.64 (P < .005) for the training and testing cohorts, respectively. CONCLUSION With the use of machine learning techniques to analyze imaging features derived from pre- and posttherapy multimodal MRI, we were able to develop a predictive model for patient OS that could potentially assist clinical decision making.
Collapse
Affiliation(s)
- Ken Chang
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts (K.C., B.Z., R.R., D.S., N.W., R.Y.H.); Department of Radiology, College of Physicians and Surgeons, Columbia University, New York, New York (X.G., M.Z., B.Z.); Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.)
| | - Biqi Zhang
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts (K.C., B.Z., R.R., D.S., N.W., R.Y.H.); Department of Radiology, College of Physicians and Surgeons, Columbia University, New York, New York (X.G., M.Z., B.Z.); Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.)
| | - Xiaotao Guo
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts (K.C., B.Z., R.R., D.S., N.W., R.Y.H.); Department of Radiology, College of Physicians and Surgeons, Columbia University, New York, New York (X.G., M.Z., B.Z.); Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.)
| | - Min Zong
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts (K.C., B.Z., R.R., D.S., N.W., R.Y.H.); Department of Radiology, College of Physicians and Surgeons, Columbia University, New York, New York (X.G., M.Z., B.Z.); Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.)
| | - Rifaquat Rahman
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts (K.C., B.Z., R.R., D.S., N.W., R.Y.H.); Department of Radiology, College of Physicians and Surgeons, Columbia University, New York, New York (X.G., M.Z., B.Z.); Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.)
| | - David Sanchez
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts (K.C., B.Z., R.R., D.S., N.W., R.Y.H.); Department of Radiology, College of Physicians and Surgeons, Columbia University, New York, New York (X.G., M.Z., B.Z.); Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.)
| | - Nicolette Winder
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts (K.C., B.Z., R.R., D.S., N.W., R.Y.H.); Department of Radiology, College of Physicians and Surgeons, Columbia University, New York, New York (X.G., M.Z., B.Z.); Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.)
| | - David A Reardon
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts (K.C., B.Z., R.R., D.S., N.W., R.Y.H.); Department of Radiology, College of Physicians and Surgeons, Columbia University, New York, New York (X.G., M.Z., B.Z.); Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.)
| | - Binsheng Zhao
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts (K.C., B.Z., R.R., D.S., N.W., R.Y.H.); Department of Radiology, College of Physicians and Surgeons, Columbia University, New York, New York (X.G., M.Z., B.Z.); Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.)
| | - Patrick Y Wen
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts (K.C., B.Z., R.R., D.S., N.W., R.Y.H.); Department of Radiology, College of Physicians and Surgeons, Columbia University, New York, New York (X.G., M.Z., B.Z.); Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.)
| | - Raymond Y Huang
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts (K.C., B.Z., R.R., D.S., N.W., R.Y.H.); Department of Radiology, College of Physicians and Surgeons, Columbia University, New York, New York (X.G., M.Z., B.Z.); Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.)
| |
Collapse
|
34
|
Gzell CE, Wheeler HR, McCloud P, Kastelan M, Back M. Small increases in enhancement on MRI may predict survival post radiotherapy in patients with glioblastoma. J Neurooncol 2016; 128:67-74. [PMID: 26879084 DOI: 10.1007/s11060-016-2074-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 02/10/2016] [Indexed: 11/30/2022]
Abstract
To assess impact of volumetric changes in tumour volume post chemoradiotherapy in glioblastoma. Patients managed with chemoradiotherapy between 2008 and 2011 were included. Patients with incomplete MRI sets were excluded. Analyses were performed on post-operative MRI, and MRIs at 1 month (M+1), 3 months (M+3), 5 months (M+5), 7 months (M+7), and 12 months (M+12) post completion of RT. RANO definitions of response were used for all techniques. Modified RANO criteria and two volumetric analysis techniques were used. The two volumetric analysis techniques involved utility of the Eclipse treatment planning software to calculate the volume of delineated tissue: surgical cavity plus all surrounding enhancement (Volumetric) versus surrounding enhancement only (Rim). Retrospective analysis of 49 patients with median survival of 18.4 months. Using Volumetric analysis the difference in MS for patients who had a <5 % increase versus ≥5 % at M+3 was 23.1 versus 15.1 months (p = 0.006), and M+5 was 26.3 versus 15.1 months (p = 0.006). For patients who were classified as progressive disease using modified RANO criteria at M+1 and M+3 there was a difference in MS compared with those who were not (M+1: 13.1 vs. 19.4 months, p = 0.017, M+3: 13.2 vs. 20.1 months, p = 0.096). An increase in the volume of cavity and enhancement of ≥5 % at M+3 and M+5 post RT was associated with reduced survival, suggesting that increases in radiological abnormality of <25 % may predict survival.
Collapse
Affiliation(s)
- Cecelia Elizabeth Gzell
- Northern Sydney Cancer Centre, Royal North Shore Hospital, Sydney, NSW, 2065, Australia. .,Northern Sydney Clinical School, Sydney University Medical School, Sydney, NSW, 2065, Australia. .,Genesis Cancer Care, Level A, 438 Victoria Street, Darlinghurst, Sydney, NSW, 2010, Australia.
| | - Helen R Wheeler
- Northern Sydney Cancer Centre, Royal North Shore Hospital, Sydney, NSW, 2065, Australia.,Northern Sydney Clinical School, Sydney University Medical School, Sydney, NSW, 2065, Australia
| | - Philip McCloud
- Northern Sydney Cancer Centre, Royal North Shore Hospital, Sydney, NSW, 2065, Australia
| | - Marina Kastelan
- Northern Sydney Cancer Centre, Royal North Shore Hospital, Sydney, NSW, 2065, Australia
| | - Michael Back
- Northern Sydney Cancer Centre, Royal North Shore Hospital, Sydney, NSW, 2065, Australia.,Northern Sydney Clinical School, Sydney University Medical School, Sydney, NSW, 2065, Australia
| |
Collapse
|
35
|
Rios Velazquez E, Meier R, Dunn Jr WD, Alexander B, Wiest R, Bauer S, Gutman DA, Reyes M, Aerts HJ. Fully automatic GBM segmentation in the TCGA-GBM dataset: Prognosis and correlation with VASARI features. Sci Rep 2015; 5:16822. [PMID: 26576732 PMCID: PMC4649540 DOI: 10.1038/srep16822] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 10/20/2015] [Indexed: 01/22/2023] Open
Abstract
Reproducible definition and quantification of imaging biomarkers is essential. We evaluated a fully automatic MR-based segmentation method by comparing it to manually defined sub-volumes by experienced radiologists in the TCGA-GBM dataset, in terms of sub-volume prognosis and association with VASARI features. MRI sets of 109 GBM patients were downloaded from the Cancer Imaging archive. GBM sub-compartments were defined manually and automatically using the Brain Tumor Image Analysis (BraTumIA). Spearman's correlation was used to evaluate the agreement with VASARI features. Prognostic significance was assessed using the C-index. Auto-segmented sub-volumes showed moderate to high agreement with manually delineated volumes (range (r): 0.4 - 0.86). Also, the auto and manual volumes showed similar correlation with VASARI features (auto r = 0.35, 0.43 and 0.36; manual r = 0.17, 0.67, 0.41, for contrast-enhancing, necrosis and edema, respectively). The auto-segmented contrast-enhancing volume and post-contrast abnormal volume showed the highest AUC (0.66, CI: 0.55-0.77 and 0.65, CI: 0.54-0.76), comparable to manually defined volumes (0.64, CI: 0.53-0.75 and 0.63, CI: 0.52-0.74, respectively). BraTumIA and manual tumor sub-compartments showed comparable performance in terms of prognosis and correlation with VASARI features. This method can enable more reproducible definition and quantification of imaging based biomarkers and has potential in high-throughput medical imaging research.
Collapse
Affiliation(s)
- Emmanuel Rios Velazquez
- Departments of Radiation Oncology and Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Raphael Meier
- Institute for Surgical Technology and Biomechanics , University of Bern, Switzerland
| | - William D. Dunn Jr
- Department of Biomedical Informatics, Emory University School of Medicine, Atlanta, GA, USA
| | - Brian Alexander
- Departments of Radiation Oncology and Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Roland Wiest
- Support Center for Advanced Neuroimaging (SCAN), Institute for Diagnostic and Interventional Neuroradiology, University Hospital Inselspital and University of Bern, Bern, Switzerland
| | - Stefan Bauer
- Institute for Surgical Technology and Biomechanics , University of Bern, Switzerland
- Support Center for Advanced Neuroimaging (SCAN), Institute for Diagnostic and Interventional Neuroradiology, University Hospital Inselspital and University of Bern, Bern, Switzerland
| | - David A. Gutman
- Department of Biomedical Informatics, Emory University School of Medicine, Atlanta, GA, USA
| | - Mauricio Reyes
- Institute for Surgical Technology and Biomechanics , University of Bern, Switzerland
| | - Hugo J.W.L. Aerts
- Departments of Radiation Oncology and Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Departments of Radiology, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Biostatistics & Computational Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
36
|
Gutman DA, Dunn WD, Grossmann P, Cooper LAD, Holder CA, Ligon KL, Alexander BM, Aerts HJWL. Somatic mutations associated with MRI-derived volumetric features in glioblastoma. Neuroradiology 2015; 57:1227-37. [PMID: 26337765 PMCID: PMC4648958 DOI: 10.1007/s00234-015-1576-7] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 08/10/2015] [Indexed: 12/16/2022]
Abstract
Introduction MR imaging can noninvasively visualize tumor phenotype characteristics at the macroscopic level. Here, we investigated whether somatic mutations are associated with and can be predicted by MRI-derived tumor imaging features of glioblastoma (GBM). Methods Seventy-six GBM patients were identified from The Cancer Imaging Archive for whom preoperative T1-contrast (T1C) and T2-FLAIR MR images were available. For each tumor, a set of volumetric imaging features and their ratios were measured, including necrosis, contrast enhancing, and edema volumes. Imaging genomics analysis assessed the association of these features with mutation status of nine genes frequently altered in adult GBM. Finally, area under the curve (AUC) analysis was conducted to evaluate the predictive performance of imaging features for mutational status. Results Our results demonstrate that MR imaging features are strongly associated with mutation status. For example, TP53-mutated tumors had significantly smaller contrast enhancing and necrosis volumes (p = 0.012 and 0.017, respectively) and RB1-mutated tumors had significantly smaller edema volumes (p = 0.015) compared to wild-type tumors. MRI volumetric features were also found to significantly predict mutational status. For example, AUC analysis results indicated that TP53, RB1, NF1, EGFR, and PDGFRA mutations could each be significantly predicted by at least one imaging feature. Conclusion MRI-derived volumetric features are significantly associated with and predictive of several cancer-relevant, drug-targetable DNA mutations in glioblastoma. These results may shed insight into unique growth characteristics of individual tumors at the macroscopic level resulting from molecular events as well as increase the use of noninvasive imaging in personalized medicine. Electronic supplementary material The online version of this article (doi:10.1007/s00234-015-1576-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- David A Gutman
- Departments of Neurology, Emory University School of Medicine, Atlanta, GA, USA.
- Biomedical Informatics, Emory University School of Medicine, 1648 Pierce Dr NE, Atlanta, GA, 30307, USA.
| | - William D Dunn
- Departments of Neurology, Emory University School of Medicine, Atlanta, GA, USA
- Biomedical Informatics, Emory University School of Medicine, 1648 Pierce Dr NE, Atlanta, GA, 30307, USA
| | - Patrick Grossmann
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Lee A D Cooper
- Biomedical Informatics, Emory University School of Medicine, 1648 Pierce Dr NE, Atlanta, GA, 30307, USA
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Chad A Holder
- Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Keith L Ligon
- Pathology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Brian M Alexander
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Hugo J W L Aerts
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Radiology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
37
|
Macyszyn L, Akbari H, Pisapia JM, Da X, Attiah M, Pigrish V, Bi Y, Pal S, Davuluri RV, Roccograndi L, Dahmane N, Martinez-Lage M, Biros G, Wolf RL, Bilello M, O'Rourke DM, Davatzikos C. Imaging patterns predict patient survival and molecular subtype in glioblastoma via machine learning techniques. Neuro Oncol 2015; 18:417-25. [PMID: 26188015 DOI: 10.1093/neuonc/nov127] [Citation(s) in RCA: 203] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 06/12/2015] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND MRI characteristics of brain gliomas have been used to predict clinical outcome and molecular tumor characteristics. However, previously reported imaging biomarkers have not been sufficiently accurate or reproducible to enter routine clinical practice and often rely on relatively simple MRI measures. The current study leverages advanced image analysis and machine learning algorithms to identify complex and reproducible imaging patterns predictive of overall survival and molecular subtype in glioblastoma (GB). METHODS One hundred five patients with GB were first used to extract approximately 60 diverse features from preoperative multiparametric MRIs. These imaging features were used by a machine learning algorithm to derive imaging predictors of patient survival and molecular subtype. Cross-validation ensured generalizability of these predictors to new patients. Subsequently, the predictors were evaluated in a prospective cohort of 29 new patients. RESULTS Survival curves yielded a hazard ratio of 10.64 for predicted long versus short survivors. The overall, 3-way (long/medium/short survival) accuracy in the prospective cohort approached 80%. Classification of patients into the 4 molecular subtypes of GB achieved 76% accuracy. CONCLUSIONS By employing machine learning techniques, we were able to demonstrate that imaging patterns are highly predictive of patient survival. Additionally, we found that GB subtypes have distinctive imaging phenotypes. These results reveal that when imaging markers related to infiltration, cell density, microvascularity, and blood-brain barrier compromise are integrated via advanced pattern analysis methods, they form very accurate predictive biomarkers. These predictive markers used solely preoperative images, hence they can significantly augment diagnosis and treatment of GB patients.
Collapse
Affiliation(s)
- Luke Macyszyn
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, Pennsylvania (L.M., J.M.P., M.A., L.R., N.D., D.M.O.); Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., R.L.W., M.B., C.D.); Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., V.P., M.B., C.D.); Department of Biomedical Informatics, Northwestern University Feinberg School of Medicine, Chicago, Illinois (Y.B., S.P., R.V.D.); Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, Texas (G.B.); Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (M.M.-L., D.M.O.)
| | - Hamed Akbari
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, Pennsylvania (L.M., J.M.P., M.A., L.R., N.D., D.M.O.); Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., R.L.W., M.B., C.D.); Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., V.P., M.B., C.D.); Department of Biomedical Informatics, Northwestern University Feinberg School of Medicine, Chicago, Illinois (Y.B., S.P., R.V.D.); Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, Texas (G.B.); Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (M.M.-L., D.M.O.)
| | - Jared M Pisapia
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, Pennsylvania (L.M., J.M.P., M.A., L.R., N.D., D.M.O.); Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., R.L.W., M.B., C.D.); Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., V.P., M.B., C.D.); Department of Biomedical Informatics, Northwestern University Feinberg School of Medicine, Chicago, Illinois (Y.B., S.P., R.V.D.); Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, Texas (G.B.); Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (M.M.-L., D.M.O.)
| | - Xiao Da
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, Pennsylvania (L.M., J.M.P., M.A., L.R., N.D., D.M.O.); Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., R.L.W., M.B., C.D.); Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., V.P., M.B., C.D.); Department of Biomedical Informatics, Northwestern University Feinberg School of Medicine, Chicago, Illinois (Y.B., S.P., R.V.D.); Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, Texas (G.B.); Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (M.M.-L., D.M.O.)
| | - Mark Attiah
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, Pennsylvania (L.M., J.M.P., M.A., L.R., N.D., D.M.O.); Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., R.L.W., M.B., C.D.); Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., V.P., M.B., C.D.); Department of Biomedical Informatics, Northwestern University Feinberg School of Medicine, Chicago, Illinois (Y.B., S.P., R.V.D.); Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, Texas (G.B.); Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (M.M.-L., D.M.O.)
| | - Vadim Pigrish
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, Pennsylvania (L.M., J.M.P., M.A., L.R., N.D., D.M.O.); Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., R.L.W., M.B., C.D.); Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., V.P., M.B., C.D.); Department of Biomedical Informatics, Northwestern University Feinberg School of Medicine, Chicago, Illinois (Y.B., S.P., R.V.D.); Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, Texas (G.B.); Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (M.M.-L., D.M.O.)
| | - Yingtao Bi
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, Pennsylvania (L.M., J.M.P., M.A., L.R., N.D., D.M.O.); Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., R.L.W., M.B., C.D.); Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., V.P., M.B., C.D.); Department of Biomedical Informatics, Northwestern University Feinberg School of Medicine, Chicago, Illinois (Y.B., S.P., R.V.D.); Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, Texas (G.B.); Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (M.M.-L., D.M.O.)
| | - Sharmistha Pal
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, Pennsylvania (L.M., J.M.P., M.A., L.R., N.D., D.M.O.); Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., R.L.W., M.B., C.D.); Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., V.P., M.B., C.D.); Department of Biomedical Informatics, Northwestern University Feinberg School of Medicine, Chicago, Illinois (Y.B., S.P., R.V.D.); Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, Texas (G.B.); Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (M.M.-L., D.M.O.)
| | - Ramana V Davuluri
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, Pennsylvania (L.M., J.M.P., M.A., L.R., N.D., D.M.O.); Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., R.L.W., M.B., C.D.); Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., V.P., M.B., C.D.); Department of Biomedical Informatics, Northwestern University Feinberg School of Medicine, Chicago, Illinois (Y.B., S.P., R.V.D.); Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, Texas (G.B.); Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (M.M.-L., D.M.O.)
| | - Laura Roccograndi
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, Pennsylvania (L.M., J.M.P., M.A., L.R., N.D., D.M.O.); Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., R.L.W., M.B., C.D.); Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., V.P., M.B., C.D.); Department of Biomedical Informatics, Northwestern University Feinberg School of Medicine, Chicago, Illinois (Y.B., S.P., R.V.D.); Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, Texas (G.B.); Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (M.M.-L., D.M.O.)
| | - Nadia Dahmane
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, Pennsylvania (L.M., J.M.P., M.A., L.R., N.D., D.M.O.); Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., R.L.W., M.B., C.D.); Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., V.P., M.B., C.D.); Department of Biomedical Informatics, Northwestern University Feinberg School of Medicine, Chicago, Illinois (Y.B., S.P., R.V.D.); Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, Texas (G.B.); Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (M.M.-L., D.M.O.)
| | - Maria Martinez-Lage
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, Pennsylvania (L.M., J.M.P., M.A., L.R., N.D., D.M.O.); Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., R.L.W., M.B., C.D.); Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., V.P., M.B., C.D.); Department of Biomedical Informatics, Northwestern University Feinberg School of Medicine, Chicago, Illinois (Y.B., S.P., R.V.D.); Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, Texas (G.B.); Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (M.M.-L., D.M.O.)
| | - George Biros
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, Pennsylvania (L.M., J.M.P., M.A., L.R., N.D., D.M.O.); Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., R.L.W., M.B., C.D.); Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., V.P., M.B., C.D.); Department of Biomedical Informatics, Northwestern University Feinberg School of Medicine, Chicago, Illinois (Y.B., S.P., R.V.D.); Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, Texas (G.B.); Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (M.M.-L., D.M.O.)
| | - Ronald L Wolf
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, Pennsylvania (L.M., J.M.P., M.A., L.R., N.D., D.M.O.); Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., R.L.W., M.B., C.D.); Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., V.P., M.B., C.D.); Department of Biomedical Informatics, Northwestern University Feinberg School of Medicine, Chicago, Illinois (Y.B., S.P., R.V.D.); Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, Texas (G.B.); Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (M.M.-L., D.M.O.)
| | - Michel Bilello
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, Pennsylvania (L.M., J.M.P., M.A., L.R., N.D., D.M.O.); Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., R.L.W., M.B., C.D.); Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., V.P., M.B., C.D.); Department of Biomedical Informatics, Northwestern University Feinberg School of Medicine, Chicago, Illinois (Y.B., S.P., R.V.D.); Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, Texas (G.B.); Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (M.M.-L., D.M.O.)
| | - Donald M O'Rourke
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, Pennsylvania (L.M., J.M.P., M.A., L.R., N.D., D.M.O.); Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., R.L.W., M.B., C.D.); Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., V.P., M.B., C.D.); Department of Biomedical Informatics, Northwestern University Feinberg School of Medicine, Chicago, Illinois (Y.B., S.P., R.V.D.); Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, Texas (G.B.); Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (M.M.-L., D.M.O.)
| | - Christos Davatzikos
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, Pennsylvania (L.M., J.M.P., M.A., L.R., N.D., D.M.O.); Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., R.L.W., M.B., C.D.); Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., V.P., M.B., C.D.); Department of Biomedical Informatics, Northwestern University Feinberg School of Medicine, Chicago, Illinois (Y.B., S.P., R.V.D.); Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, Texas (G.B.); Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (M.M.-L., D.M.O.)
| |
Collapse
|
38
|
Scribner E, Saut O, Province P, Bag A, Colin T, Fathallah-Shaykh HM. Effects of anti-angiogenesis on glioblastoma growth and migration: model to clinical predictions. PLoS One 2014; 9:e115018. [PMID: 25506702 PMCID: PMC4266618 DOI: 10.1371/journal.pone.0115018] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 11/17/2014] [Indexed: 01/09/2023] Open
Abstract
Glioblastoma multiforme (GBM) causes significant neurological morbidity and short survival times. Brain invasion by GBM is associated with poor prognosis. Recent clinical trials of bevacizumab in newly-diagnosed GBM found no beneficial effects on overall survival times; however, the baseline health-related quality of life and performance status were maintained longer in the bevacizumab group and the glucocorticoid requirement was lower. Here, we construct a clinical-scale model of GBM whose predictions uncover a new pattern of recurrence in 11/70 bevacizumab-treated patients. The findings support an exception to the Folkman hypothesis: GBM grows in the absence of angiogenesis by a cycle of proliferation and brain invasion that expands necrosis. Furthermore, necrosis is positively correlated with brain invasion in 26 newly-diagnosed GBM. The unintuitive results explain the unusual clinical effects of bevacizumab and suggest new hypotheses on the dynamic clinical effects of migration by active transport, a mechanism of hypoxia-driven brain invasion.
Collapse
Affiliation(s)
- Elizabeth Scribner
- Department of Mathematics, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Olivier Saut
- Department of Mathematics, University of Bordeaux, Talence, France
| | - Paula Province
- Department of Neurology, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Asim Bag
- Department of Radiology, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Thierry Colin
- Department of Mathematics, University of Bordeaux, Talence, France
| | - Hassan M. Fathallah-Shaykh
- Department of Mathematics, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Department of Neurology, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- * E-mail:
| |
Collapse
|