1
|
di Noia C, Grist JT, Riemer F, Lyasheva M, Fabozzi M, Castelli M, Lodi R, Tonon C, Rundo L, Zaccagna F. Predicting Survival in Patients with Brain Tumors: Current State-of-the-Art of AI Methods Applied to MRI. Diagnostics (Basel) 2022; 12:diagnostics12092125. [PMID: 36140526 PMCID: PMC9497964 DOI: 10.3390/diagnostics12092125] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/05/2022] [Accepted: 08/17/2022] [Indexed: 11/24/2022] Open
Abstract
Given growing clinical needs, in recent years Artificial Intelligence (AI) techniques have increasingly been used to define the best approaches for survival assessment and prediction in patients with brain tumors. Advances in computational resources, and the collection of (mainly) public databases, have promoted this rapid development. This narrative review of the current state-of-the-art aimed to survey current applications of AI in predicting survival in patients with brain tumors, with a focus on Magnetic Resonance Imaging (MRI). An extensive search was performed on PubMed and Google Scholar using a Boolean research query based on MeSH terms and restricting the search to the period between 2012 and 2022. Fifty studies were selected, mainly based on Machine Learning (ML), Deep Learning (DL), radiomics-based methods, and methods that exploit traditional imaging techniques for survival assessment. In addition, we focused on two distinct tasks related to survival assessment: the first on the classification of subjects into survival classes (short and long-term or eventually short, mid and long-term) to stratify patients in distinct groups. The second focused on quantification, in days or months, of the individual survival interval. Our survey showed excellent state-of-the-art methods for the first, with accuracy up to ∼98%. The latter task appears to be the most challenging, but state-of-the-art techniques showed promising results, albeit with limitations, with C-Index up to ∼0.91. In conclusion, according to the specific task, the available computational methods perform differently, and the choice of the best one to use is non-univocal and dependent on many aspects. Unequivocally, the use of features derived from quantitative imaging has been shown to be advantageous for AI applications, including survival prediction. This evidence from the literature motivates further research in the field of AI-powered methods for survival prediction in patients with brain tumors, in particular, using the wealth of information provided by quantitative MRI techniques.
Collapse
Affiliation(s)
- Christian di Noia
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum—University of Bologna, 40125 Bologna, Italy
| | - James T. Grist
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford OX1 3PT, UK
- Department of Radiology, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK
- Oxford Centre for Clinical Magnetic Research Imaging, University of Oxford, Oxford OX3 9DU, UK
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham B15 2SY, UK
| | - Frank Riemer
- Mohn Medical Imaging and Visualization Centre (MMIV), Department of Radiology, Haukeland University Hospital, N-5021 Bergen, Norway
| | - Maria Lyasheva
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Miriana Fabozzi
- Centro Medico Polispecialistico (CMO), 80058 Torre Annunziata, Italy
| | - Mauro Castelli
- NOVA Information Management School (NOVA IMS), Universidade NOVA de Lisboa, Campus de Campolide, 1070-312 Lisboa, Portugal
| | - Raffaele Lodi
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum—University of Bologna, 40125 Bologna, Italy
- Functional and Molecular Neuroimaging Unit, IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy
| | - Caterina Tonon
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum—University of Bologna, 40125 Bologna, Italy
- Functional and Molecular Neuroimaging Unit, IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy
| | - Leonardo Rundo
- Department of Information and Electrical Engineering and Applied Mathematics, University of Salerno, 84084 Fisciano, Italy
| | - Fulvio Zaccagna
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum—University of Bologna, 40125 Bologna, Italy
- Functional and Molecular Neuroimaging Unit, IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy
- Correspondence: ; Tel.: +39-0514969951
| |
Collapse
|
2
|
Zhou Q, Xue C, Ke X, Zhou J. Treatment Response and Prognosis Evaluation in High-Grade Glioma: An Imaging Review Based on MRI. J Magn Reson Imaging 2022; 56:325-340. [PMID: 35129845 DOI: 10.1002/jmri.28103] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/25/2022] [Accepted: 01/25/2022] [Indexed: 12/19/2022] Open
Abstract
In recent years, the development of advanced magnetic resonance imaging (MRI) technology and machine learning (ML) have created new tools for evaluating treatment response and prognosis of patients with high-grade gliomas (HGG); however, patient prognosis has not improved significantly. This is mainly due to the heterogeneity between and within HGG tumors, resulting in standard treatment methods not benefitting all patients. Moreover, the survival of patients with HGG is not only related to tumor cells, but also to noncancer cells in the tumor microenvironment (TME). Therefore, during preoperative diagnosis and follow-up treatment of patients with HGG, noninvasive imaging markers are needed to characterize intratumoral heterogeneity, and then to evaluate treatment response and predict prognosis, timeously adjust treatment strategies, and achieve individualized diagnosis and treatment. In this review, we summarize the research progress of conventional MRI, advanced MRI technology, and ML in evaluation of treatment response and prognosis of patients with HGG. We further discuss the significance of the TME in the prognosis of HGG patients, associate imaging features with the TME, indirectly reflecting the heterogeneity within the tumor, and shifting treatment strategies from tumor cells alone to systemic therapy of the TME, which may be a major development direction in the future. LEVEL OF EVIDENCE: 5 TECHNICAL EFFICACY STAGE: 4.
Collapse
Affiliation(s)
- Qing Zhou
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, Gansu, China.,Second Clinical School, Lanzhou University, Lanzhou, Gansu, China.,Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, Gansu, China.,Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | - Caiqiang Xue
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, Gansu, China.,Second Clinical School, Lanzhou University, Lanzhou, Gansu, China.,Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, Gansu, China.,Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | - Xiaoai Ke
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, Gansu, China.,Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, Gansu, China.,Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | - Junlin Zhou
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, Gansu, China.,Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, Gansu, China.,Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| |
Collapse
|
3
|
Wang D, Liu C, Wang X, Liu X, Lan C, Zhao P, Cho WC, Graeber MB, Liu Y. Automated Machine-Learning Framework Integrating Histopathological and Radiological Information for Predicting IDH1 Mutation Status in Glioma. FRONTIERS IN BIOINFORMATICS 2021; 1:718697. [PMID: 36303770 PMCID: PMC9581043 DOI: 10.3389/fbinf.2021.718697] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 09/28/2021] [Indexed: 09/01/2023] Open
Abstract
Diffuse gliomas are the most common malignant primary brain tumors. Identification of isocitrate dehydrogenase 1 (IDH1) mutations aids the diagnostic classification of these tumors and the prediction of their clinical outcomes. While histology continues to play a key role in frozen section diagnosis, as a diagnostic reference and as a method for monitoring disease progression, recent research has demonstrated the ability of multi-parametric magnetic resonance imaging (MRI) sequences for predicting IDH genotypes. In this paper, we aim to improve the prediction accuracy of IDH1 genotypes by integrating multi-modal imaging information from digitized histopathological data derived from routine histological slide scans and the MRI sequences including T1-contrast (T1) and Fluid-attenuated inversion recovery imaging (T2-FLAIR). In this research, we have established an automated framework to process, analyze and integrate the histopathological and radiological information from high-resolution pathology slides and multi-sequence MRI scans. Our machine-learning framework comprehensively computed multi-level information including molecular level, cellular level, and texture level information to reflect predictive IDH genotypes. Firstly, an automated pre-processing was developed to select the regions of interest (ROIs) from pathology slides. Secondly, to interactively fuse the multimodal complementary information, comprehensive feature information was extracted from the pathology ROIs and segmented tumor regions (enhanced tumor, edema and non-enhanced tumor) from MRI sequences. Thirdly, a Random Forest (RF)-based algorithm was employed to identify and quantitatively characterize histopathological and radiological imaging origins, respectively. Finally, we integrated multi-modal imaging features with a machine-learning algorithm and tested the performance of the framework for IDH1 genotyping, we also provided visual and statistical explanation to support the understanding on prediction outcomes. The training and testing experiments on 217 pathologically verified IDH1 genotyped glioma cases from multi-resource validated that our fully automated machine-learning model predicted IDH1 genotypes with greater accuracy and reliability than models that were based on radiological imaging data only. The accuracy of IDH1 genotype prediction was 0.90 compared to 0.82 for radiomic result. Thus, the integration of multi-parametric imaging features for automated analysis of cross-modal biomedical data improved the prediction accuracy of glioma IDH1 genotypes.
Collapse
Affiliation(s)
- Dingqian Wang
- School of Computer Science, The University of Sydney, Sydney, NSW, Australia
| | - Cuicui Liu
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xiuying Wang
- School of Computer Science, The University of Sydney, Sydney, NSW, Australia
| | - Xuejun Liu
- Department of Radiology, Hospital Affiliated to Qingdao University, Qingdao, China
| | - Chuanjin Lan
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Peng Zhao
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - William C. Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong, SAR China
| | - Manuel B. Graeber
- Ken Parker Brain Tumor Research Laboratories, Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Yingchao Liu
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| |
Collapse
|
4
|
Auer TA, Della Seta M, Collettini F, Chapiro J, Zschaeck S, Ghadjar P, Badakhshi H, Florange J, Hamm B, Budach V, Kaul D. Quantitative volumetric assessment of baseline enhancing tumor volume as an imaging biomarker predicts overall survival in patients with glioblastoma. Acta Radiol 2021; 62:1200-1207. [PMID: 32938221 DOI: 10.1177/0284185120953796] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is the commonest malignant primary brain tumor and still has one of the worst prognoses among cancers in general. There is a need for non-invasive methods to predict individual prognosis in patients with GBM. PURPOSE To evaluate quantitative volumetric tissue assessment of enhancing tumor volume on cranial magnetic resonance imaging (MRI) as an imaging biomarker for predicting overall survival (OS) in patients with GBM. MATERIAL AND METHODS MRI scans of 49 patients with histopathologically confirmed GBM were analyzed retrospectively. Baseline contrast-enhanced (CE) MRI sequences were transferred to a segmentation-based three-dimensional quantification tool, and the enhancing tumor component was analyzed. Based on a cut-off percentage of the enhancing tumor volume (PoETV) of >84.78%, samples were dichotomized, and the OS and intracranial progression-free survival (PFS) were evaluated. Univariable and multivariable analyses, including variables such as sex, Karnofsky Performance Status score, O6-methylguanine-DNA-methyltransferase status, age, and resection status, were performed using the Cox regression model. RESULTS The median OS and PFS were 16.9 and 7 months in the entire cohort, respectively. Patients with a CE tumor volume of >84.78% showed a significantly shortened OS (12.9 months) compared to those with a CE tumor volume of ≤84.78% (17.7 months) (hazard ratio [HR] 2.72; 95% confidence interval [CI] 1.22-6.03; P = 0.01). Multivariable analysis confirmed that PoETV had a significant prognostic role (HR 2.47; 95% CI 1.08-5.65; P = 0.03). CONCLUSION We observed a correlation between PoETV and OS. This imaging biomarker may help predict the OS of patients with GBM.
Collapse
Affiliation(s)
- Timo A Auer
- Department of Radiology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Marta Della Seta
- Department of Radiology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Federico Collettini
- Department of Radiology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Julius Chapiro
- Department of Radiology, Yale University, New Haven, CT, USA
| | - Sebastian Zschaeck
- Department of Radiation Oncology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Pirus Ghadjar
- Department of Radiation Oncology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Harun Badakhshi
- Department of Radiation Oncology, Ernst von Bergmann Medical Center, Potsdam, Germany
| | - Julian Florange
- Department of Radiation Oncology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Bernd Hamm
- Department of Radiology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Volker Budach
- Department of Radiation Oncology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - David Kaul
- Department of Radiation Oncology, Charité Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
5
|
Tewarie IA, Senders JT, Kremer S, Devi S, Gormley WB, Arnaout O, Smith TR, Broekman MLD. Survival prediction of glioblastoma patients-are we there yet? A systematic review of prognostic modeling for glioblastoma and its clinical potential. Neurosurg Rev 2021; 44:2047-2057. [PMID: 33156423 PMCID: PMC8338817 DOI: 10.1007/s10143-020-01430-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 09/28/2020] [Accepted: 10/27/2020] [Indexed: 02/07/2023]
Abstract
Glioblastoma is associated with a poor prognosis. Even though survival statistics are well-described at the population level, it remains challenging to predict the prognosis of an individual patient despite the increasing number of prognostic models. The aim of this study is to systematically review the literature on prognostic modeling in glioblastoma patients. A systematic literature search was performed to identify all relevant studies that developed a prognostic model for predicting overall survival in glioblastoma patients following the PRISMA guidelines. Participants, type of input, algorithm type, validation, and testing procedures were reviewed per prognostic model. Among 595 citations, 27 studies were included for qualitative review. The included studies developed and evaluated a total of 59 models, of which only seven were externally validated in a different patient cohort. The predictive performance among these studies varied widely according to the AUC (0.58-0.98), accuracy (0.69-0.98), and C-index (0.66-0.70). Three studies deployed their model as an online prediction tool, all of which were based on a statistical algorithm. The increasing performance of survival prediction models will aid personalized clinical decision-making in glioblastoma patients. The scientific realm is gravitating towards the use of machine learning models developed on high-dimensional data, often with promising results. However, none of these models has been implemented into clinical care. To facilitate the clinical implementation of high-performing survival prediction models, future efforts should focus on harmonizing data acquisition methods, improving model interpretability, and externally validating these models in multicentered, prospective fashion.
Collapse
Affiliation(s)
- Ishaan Ashwini Tewarie
- Department of Neurosurgery, Haaglanden Medical Center, Lijnbaan 32, 2512 VA, The Hague, The Netherlands
- Faculty of Medicine, Erasmus University Rotterdam, Rotterdam, The Netherlands
- Computational Neurosciences Outcomes Center, Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Joeky T Senders
- Department of Neurosurgery, Haaglanden Medical Center, Lijnbaan 32, 2512 VA, The Hague, The Netherlands
- Computational Neurosciences Outcomes Center, Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Stijn Kremer
- Department of Neurosurgery, Haaglanden Medical Center, Lijnbaan 32, 2512 VA, The Hague, The Netherlands
| | - Sharmila Devi
- Computational Neurosciences Outcomes Center, Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- King's College, London, UK
| | - William B Gormley
- Computational Neurosciences Outcomes Center, Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Omar Arnaout
- Computational Neurosciences Outcomes Center, Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Timothy R Smith
- Computational Neurosciences Outcomes Center, Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Marike L D Broekman
- Department of Neurosurgery, Haaglanden Medical Center, Lijnbaan 32, 2512 VA, The Hague, The Netherlands.
- Computational Neurosciences Outcomes Center, Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Department of Neurosurgery, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
6
|
Buchlak QD, Esmaili N, Leveque JC, Bennett C, Farrokhi F, Piccardi M. Machine learning applications to neuroimaging for glioma detection and classification: An artificial intelligence augmented systematic review. J Clin Neurosci 2021; 89:177-198. [PMID: 34119265 DOI: 10.1016/j.jocn.2021.04.043] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 04/30/2021] [Indexed: 12/13/2022]
Abstract
Glioma is the most common primary intraparenchymal tumor of the brain and the 5-year survival rate of high-grade glioma is poor. Magnetic resonance imaging (MRI) is essential for detecting, characterizing and monitoring brain tumors but definitive diagnosis still relies on surgical pathology. Machine learning has been applied to the analysis of MRI data in glioma research and has the potential to change clinical practice and improve patient outcomes. This systematic review synthesizes and analyzes the current state of machine learning applications to glioma MRI data and explores the use of machine learning for systematic review automation. Various datapoints were extracted from the 153 studies that met inclusion criteria and analyzed. Natural language processing (NLP) analysis involved keyword extraction, topic modeling and document classification. Machine learning has been applied to tumor grading and diagnosis, tumor segmentation, non-invasive genomic biomarker identification, detection of progression and patient survival prediction. Model performance was generally strong (AUC = 0.87 ± 0.09; sensitivity = 0.87 ± 0.10; specificity = 0.0.86 ± 0.10; precision = 0.88 ± 0.11). Convolutional neural network, support vector machine and random forest algorithms were top performers. Deep learning document classifiers yielded acceptable performance (mean 5-fold cross-validation AUC = 0.71). Machine learning tools and data resources were synthesized and summarized to facilitate future research. Machine learning has been widely applied to the processing of MRI data in glioma research and has demonstrated substantial utility. NLP and transfer learning resources enabled the successful development of a replicable method for automating the systematic review article screening process, which has potential for shortening the time from discovery to clinical application in medicine.
Collapse
Affiliation(s)
- Quinlan D Buchlak
- School of Medicine, The University of Notre Dame Australia, Sydney, NSW, Australia.
| | - Nazanin Esmaili
- School of Medicine, The University of Notre Dame Australia, Sydney, NSW, Australia; Faculty of Engineering and IT, University of Technology Sydney, Ultimo, NSW, Australia
| | | | - Christine Bennett
- School of Medicine, The University of Notre Dame Australia, Sydney, NSW, Australia
| | - Farrokh Farrokhi
- Neuroscience Institute, Virginia Mason Medical Center, Seattle, WA, USA
| | - Massimo Piccardi
- Faculty of Engineering and IT, University of Technology Sydney, Ultimo, NSW, Australia
| |
Collapse
|
7
|
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
|
8
|
Ebrahimi Zade A, Shahabi Haghighi S, Soltani M. A neuro evolutionary algorithm for patient calibrated prediction of survival in Glioblastoma patients. J Biomed Inform 2021; 115:103694. [PMID: 33545332 DOI: 10.1016/j.jbi.2021.103694] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 01/28/2021] [Accepted: 01/29/2021] [Indexed: 01/30/2023]
Abstract
BACKGROUND AND OBJECTIVES Glioblastoma multiforme (GBM) is the most common and malignant type of primary brain tumors. Radiation therapy (RT) plus concomitant and adjuvant Temozolomide (TMZ) constitute standard treatment of GBM. Existing models for GBM growth do not consider the effect of different schedules on tumor growth and patient survival. However, clinical trials show that treatment schedule and drug dosage significantly affect patient survival. The goal is to provide a patient calibrated model for predicting survival according to the treatment schedule. METHODS We propose a top-down method based on artificial neural networks (ANN) and genetic algorithm (GA) to predict survival of GBM patients. A feed forward undercomplete Autoencoder network is integrated with the neuro-evolutionary (NE) algorithm in order to extract a compressed representation of input clinical data. The proposed NE algorithm uses GA to obtain optimal architecture of a multi-layer perceptron (MLP). Taguchi L16 orthogonal design of experiments is used to tune parameters of the proposed NE algorithm. Finally, the optimal MLP is used to predict survival of GBM patients. RESULTS Data from 8 related clinical trials have been collected and integrated to train the model. From 847 evaluable cases, 719 were used for train and validation and the remaining 128 cases were used to test the model. Mean absolute error of the predictions on the test data is 0.087 months which shows excellent performance of the proposed model in predicting survival of the patients. Also, the results show that the proposed NE algorithm is superior to other existing models in both the mean and variability of the prediction error.
Collapse
Affiliation(s)
- Amir Ebrahimi Zade
- Faculty of Industrial Engineering and Systems Management, Amirkabir University of Technology, Tehran, Iran
| | | | - M Soltani
- Faculty of Mechanical Engineering, K.N. Toosi University of Technology, Tehran, Iran; Advanced Bioengineering Initiative Center, Computational Medicine Center, K. N. Toosi University of Technology, Tehran, Iran; Centre for Biotechnology and Bioengineering (CBB), University of Waterloo, Waterloo, ON, Canada; Department of Electrical and Computer Engineering, University of Waterloo, Waterloo, ON, Canada
| |
Collapse
|
9
|
Evaluating survival in subjects with astrocytic brain tumors by dynamic susceptibility-weighted perfusion MR imaging. PLoS One 2021; 16:e0244275. [PMID: 33406116 PMCID: PMC7787526 DOI: 10.1371/journal.pone.0244275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 12/07/2020] [Indexed: 12/02/2022] Open
Abstract
Purpose Studies have evaluated the application of perfusion MR for predicting survival in patients with astrocytic brain tumors, but few of them statistically adjust their results to reflect the impact of the variability of treatment administered in the patients. Our aim was to analyze the association between the perfusion values and overall survival time, with adjustment for various clinical factors, including initial treatments and follow-up treatments. Materials and methods This study consisted of 51 patients with astrocytic brain tumors who underwent perfusion-weighted MRI with MultiHance® at a dose of 0.1 mmol/kg prior to initial surgery. We measured the mean rCBV, the 5% & 10% maximum rCBV, and the variation of rCBV in the tumors. Comparisons were made between patients with and without 2-year survival using two-sample t-test or Wilcoxon rank-sum test for the continuous data, or chi-square and Fisher exact tests for categorical data. The multivariate cox-proportional hazard regression was fit to evaluate the association between rCBV and overall survival time, with adjustment for clinical factors. Results Patients who survived less than 2 years after diagnosis had a higher mean and maximum rCBV and a larger variation of rCBV. After adjusting for clinical factors including therapeutic measures, we found no significant association of overall survival time within 2 years with any of these rCBV values. Conclusions Although patients who survived less than 2 years had a higher mean and maximum rCBV and a larger variation of rCBV, rCBV itself may not be used independently for predicting 2-year survival of patients with astrocytic brain tumors.
Collapse
|
10
|
Hussain MM, Shabbir A, Bakhshi SK, Shamim MS. Are Thinking Machines Breaking New Frontiers in Neuro-Oncology? A Narrative Review on the Emerging Role of Machine Learning in Neuro-Oncological Practice. Asian J Neurosurg 2021; 16:8-13. [PMID: 34211861 PMCID: PMC8202358 DOI: 10.4103/ajns.ajns_265_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/07/2020] [Accepted: 09/17/2020] [Indexed: 11/21/2022] Open
Abstract
Medical science in general and oncology in particular are dynamic, rapidly evolving subjects. Brain and spine tumors, whether primary or secondary, constitute a significant number of cases in any oncological practice. With the rapid influx of data in all aspects of neuro-oncological care, it is almost impossible for practicing clinicians to remain abreast with the current trends, or to synthesize the available data for it to be maximally beneficial for their patients. Machine-learning (ML) tools are fast gaining acceptance as an alternative to conventional reliance on online data. ML uses artificial intelligence to provide a computer algorithm-based information to clinicians. Different ML models have been proposed in the literature with a variable degree of precision and database requirements. ML can potentially solve the aforementioned problems for practicing clinicians by not just extracting and analyzing useful data, by minimizing or eliminating certain potential areas of human error, by creating patient-specific treatment plans, and also by predicting outcomes with reasonable accuracy. Current information on ML in neuro-oncology is scattered, and this literature review is an attempt to consolidate it and provide recent updates.
Collapse
Affiliation(s)
| | - Ainsia Shabbir
- Department of Computer and Information Systems Engineering, NED University of Engineering and Technology, Karachi, Pakistan
| | | | | |
Collapse
|
11
|
Predicting Survival in Glioblastoma Patients Using Diffusion MR Imaging Metrics-A Systematic Review. Cancers (Basel) 2020; 12:cancers12102858. [PMID: 33020420 PMCID: PMC7600641 DOI: 10.3390/cancers12102858] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 09/28/2020] [Accepted: 10/01/2020] [Indexed: 12/20/2022] Open
Abstract
Simple Summary An accurate survival analysis is crucial for disease management in glioblastoma (GBM) patients. Due to the ability of the diffusion MRI techniques of providing a quantitative assessment of GBM tumours, an ever-growing number of studies aimed at investigating the role of diffusion MRI metrics in survival prediction of GBM patients. Since the role of diffusion MRI in prediction and evaluation of survival outcomes has not been fully addressed and results are often controversial or unsatisfactory, we performed this systematic review in order to collect, summarize and evaluate all studies evaluating the role of diffusion MRI metrics in predicting survival in GBM patients. We found that quantitative diffusion MRI metrics provide useful information for predicting survival outcomes in GBM patients, mainly in combination with other clinical and multimodality imaging parameters. Abstract Despite advances in surgical and medical treatment of glioblastoma (GBM), the medium survival is about 15 months and varies significantly, with occasional longer survivors and individuals whose tumours show a significant response to therapy with respect to others. Diffusion MRI can provide a quantitative assessment of the intratumoral heterogeneity of GBM infiltration, which is of clinical significance for targeted surgery and therapy, and aimed at improving GBM patient survival. So, the aim of this systematic review is to assess the role of diffusion MRI metrics in predicting survival of patients with GBM. According to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement, a systematic literature search was performed to identify original articles since 2010 that evaluated the association of diffusion MRI metrics with overall survival (OS) and progression-free survival (PFS). The quality of the included studies was evaluated using the QUIPS tool. A total of 52 articles were selected. The most examined metrics were associated with the standard Diffusion Weighted Imaging (DWI) (34 studies) and Diffusion Tensor Imaging (DTI) models (17 studies). Our findings showed that quantitative diffusion MRI metrics provide useful information for predicting survival outcomes in GBM patients, mainly in combination with other clinical and multimodality imaging parameters.
Collapse
|
12
|
Zhuge Y, Ning H, Mathen P, Cheng JY, Krauze AV, Camphausen K, Miller RW. Automated glioma grading on conventional MRI images using deep convolutional neural networks. Med Phys 2020; 47:3044-3053. [PMID: 32277478 PMCID: PMC8494136 DOI: 10.1002/mp.14168] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 03/09/2020] [Accepted: 03/25/2020] [Indexed: 01/05/2023] Open
Abstract
PURPOSE Gliomas are the most common primary tumor of the brain and are classified into grades I-IV of the World Health Organization (WHO), based on their invasively histological appearance. Gliomas grading plays an important role to determine the treatment plan and prognosis prediction. In this study we propose two novel methods for automatic, non-invasively distinguishing low-grade (Grades II and III) glioma (LGG) and high-grade (grade IV) glioma (HGG) on conventional MRI images by using deep convolutional neural networks (CNNs). METHODS All MRI images have been preprocessed first by rigid image registration and intensity inhomogeneity correction. Both proposed methods consist of two steps: (a) three-dimensional (3D) brain tumor segmentation based on a modification of the popular U-Net model; (b) tumor classification on segmented brain tumor. In the first method, the slice with largest area of tumor is determined and the state-of-the-art mask R-CNN model is employed for tumor grading. To improve the performance of the grading model, a two-dimensional (2D) data augmentation has been implemented to increase both the amount and the diversity of the training images. In the second method, denoted as 3DConvNet, a 3D volumetric CNNs is applied directly on bounding image regions of segmented tumor for classification, which can fully leverage the 3D spatial contextual information of volumetric image data. RESULTS The proposed schemes were evaluated on The Cancer Imaging Archive (TCIA) low grade glioma (LGG) data, and the Multimodal Brain Tumor Image Segmentation (BraTS) Benchmark 2018 training datasets with fivefold cross validation. All data are divided into training, validation, and test sets. Based on biopsy-proven ground truth, the performance metrics of sensitivity, specificity, and accuracy are measured on the test sets. The results are 0.935 (sensitivity), 0.972 (specificity), and 0.963 (accuracy) for the 2D Mask R-CNN based method, and 0.947 (sensitivity), 0.968 (specificity), and 0.971 (accuracy) for the 3DConvNet method, respectively. In regard to efficiency, for 3D brain tumor segmentation, the program takes around ten and a half hours for training with 300 epochs on BraTS 2018 dataset and takes only around 50 s for testing of a typical image with a size of 160 × 216 × 176. For 2D Mask R-CNN based tumor grading, the program takes around 4 h for training with around 60 000 iterations, and around 1 s for testing of a 2D slice image with size of 128 × 128. For 3DConvNet based tumor grading, the program takes around 2 h for training with 10 000 iterations, and 0.25 s for testing of a 3D cropped image with size of 64 × 64 × 64, using a DELL PRECISION Tower T7910, with two NVIDIA Titan Xp GPUs. CONCLUSIONS Two effective glioma grading methods on conventional MRI images using deep convolutional neural networks have been developed. Our methods are fully automated without manual specification of region-of-interests and selection of slices for model training, which are common in traditional machine learning based brain tumor grading methods. This methodology may play a crucial role in selecting effective treatment options and survival predictions without the need for surgical biopsy.
Collapse
Affiliation(s)
- Ying Zhuge
- Radiation Oncology Branch, National Cancer Institute National Institutes of Health, Bethesda, MD 20892, USA
| | - Holly Ning
- Radiation Oncology Branch, National Cancer Institute National Institutes of Health, Bethesda, MD 20892, USA
| | - Peter Mathen
- Radiation Oncology Branch, National Cancer Institute National Institutes of Health, Bethesda, MD 20892, USA
| | - Jason Y. Cheng
- Radiation Oncology Branch, National Cancer Institute National Institutes of Health, Bethesda, MD 20892, USA
| | - Andra V. Krauze
- Division of Radiation Oncology and Developmental Radiotherapeutics, BC Cancer, Vancouver, BC, Canada
| | - Kevin Camphausen
- Radiation Oncology Branch, National Cancer Institute National Institutes of Health, Bethesda, MD 20892, USA
| | - Robert W. Miller
- Radiation Oncology Branch, National Cancer Institute National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
13
|
Ius T, Pignotti F, Della Pepa GM, La Rocca G, Somma T, Isola M, Battistella C, Gaudino S, Polano M, Dal Bo M, Bagatto D, Pegolo E, Chiesa S, Arcicasa M, Olivi A, Skrap M, Sabatino G. A Novel Comprehensive Clinical Stratification Model to Refine Prognosis of Glioblastoma Patients Undergoing Surgical Resection. Cancers (Basel) 2020; 12:E386. [PMID: 32046132 PMCID: PMC7072471 DOI: 10.3390/cancers12020386] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/29/2020] [Accepted: 02/05/2020] [Indexed: 12/14/2022] Open
Abstract
Despite recent discoveries in genetics and molecular fields, glioblastoma (GBM) prognosis still remains unfavorable with less than 10% of patients alive 5 years after diagnosis. Numerous studies have focused on the research of biological biomarkers to stratify GBM patients. We addressed this issue in our study by using clinical/molecular and image data, which is generally available to Neurosurgical Departments in order to create a prognostic score that can be useful to stratify GBM patients undergoing surgical resection. By using the random forest approach [CART analysis (classification and regression tree)] on Survival time data of 465 cases, we developed a new prediction score resulting in 10 groups based on extent of resection (EOR), age, tumor volumetric features, intraoperative protocols and tumor molecular classes. The resulting tree was trimmed according to similarities in the relative hazard ratios amongst groups, giving rise to a 5-group classification tree. These 5 groups were different in terms of overall survival (OS) (p < 0.000). The score performance in predicting death was defined by a Harrell's c-index of 0.79 (95% confidence interval [0.76-0.81]). The proposed score could be useful in a clinical setting to refine the prognosis of GBM patients after surgery and prior to postoperative treatment.
Collapse
Affiliation(s)
- Tamara Ius
- Neurosurgery Unit, Department of Neuroscience, Santa Maria della Misericordia University Hospital, 33100 Udine, Italy;
| | - Fabrizio Pignotti
- Department of Neurosurgery, Mater Olbia Hospital, 07026 Olbia, Italy; (F.P.); (G.S.); (G.L.R.)
| | | | - Giuseppe La Rocca
- Department of Neurosurgery, Mater Olbia Hospital, 07026 Olbia, Italy; (F.P.); (G.S.); (G.L.R.)
- Institute of Neurosurgery, Catholic University, 00168 Rome, Italy; (G.M.D.P.); (A.O.)
| | - Teresa Somma
- Division of Neurosurgery, Department of Neurosciences, Reproductive and Odontostomatological Sciences, Università degli Studi di Napoli Federico II, 80131 Naples, Italy;
| | - Miriam Isola
- Department of Medicine, Santa Maria della Misericordia University Hospital, 33100 Udine, Italy; (M.I.); (C.B.)
| | - Claudio Battistella
- Department of Medicine, Santa Maria della Misericordia University Hospital, 33100 Udine, Italy; (M.I.); (C.B.)
| | - Simona Gaudino
- Institute of radiology, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy;
| | - Maurizio Polano
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (M.P.); (M.D.B.)
| | - Michele Dal Bo
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (M.P.); (M.D.B.)
| | - Daniele Bagatto
- Neuroradiology Unit, Department of Diagnostic Imaging ASUIUD Udine, 33100 Udine, Italy;
| | - Enrico Pegolo
- Institute of Pathology, Santa Maria della Misericordia University Hospital, 33100 Udine, Italy;
| | - Silvia Chiesa
- Radiation Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy;
| | - Mauro Arcicasa
- Department of Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy;
| | - Alessandro Olivi
- Institute of Neurosurgery, Catholic University, 00168 Rome, Italy; (G.M.D.P.); (A.O.)
| | - Miran Skrap
- Neurosurgery Unit, Department of Neuroscience, Santa Maria della Misericordia University Hospital, 33100 Udine, Italy;
| | - Giovanni Sabatino
- Department of Neurosurgery, Mater Olbia Hospital, 07026 Olbia, Italy; (F.P.); (G.S.); (G.L.R.)
- Institute of Neurosurgery, Catholic University, 00168 Rome, Italy; (G.M.D.P.); (A.O.)
| |
Collapse
|
14
|
Liu L, Zhang H, Wu J, Yu Z, Chen X, Rekik I, Wang Q, Lu J, Shen D. Overall survival time prediction for high-grade glioma patients based on large-scale brain functional networks. Brain Imaging Behav 2020; 13:1333-1351. [PMID: 30155788 DOI: 10.1007/s11682-018-9949-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
High-grade glioma (HGG) is a lethal cancer with poor outcome. Accurate preoperative overall survival (OS) time prediction for HGG patients is crucial for treatment planning. Traditional presurgical and noninvasive OS prediction studies have used radiomics features at the local lesion area based on the magnetic resonance images (MRI). However, the highly complex lesion MRI appearance may have large individual variability, which could impede accurate individualized OS prediction. In this paper, we propose a novel concept, namely brain connectomics-based OS prediction. It is based on presurgical resting-state functional MRI (rs-fMRI) and the non-local, large-scale brain functional networks where the global and systemic prognostic features rather than the local lesion appearance are used to predict OS. We propose that the connectomics features could capture tumor-induced network-level alterations that are associated with prognosis. We construct both low-order (by means of sparse representation with regional rs-fMRI signals) and high-order functional connectivity (FC) networks (characterizing more complex multi-regional relationship by synchronized dynamics FC time courses). Then, we conduct a graph-theoretic analysis on both networks for a jointly, machine-learning-based individualized OS prediction. Based on a preliminary dataset (N = 34 with bad OS, mean OS, ~400 days; N = 34 with good OS, mean OS, ~1030 days), we achieve a promising OS prediction accuracy (86.8%) on separating the individuals with bad OS from those with good OS. However, if using only conventionally derived descriptive features (e.g., age and tumor characteristics), the accuracy is low (63.2%). Our study highlights the importance of the rs-fMRI and brain functional connectomics for treatment planning.
Collapse
Affiliation(s)
- Luyan Liu
- Med-X Research Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai, 200030, China.,Department of Radiology and BRIC, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Han Zhang
- Department of Radiology and BRIC, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jinsong Wu
- Glioma Surgery Division, Neurosurgery Department of Huashan Hospital, Fudan University, Shanghai, 200040, China.,Shanghai Key Lab of Medical Image Computing and Computer-Assisted Intervention, Shanghai, 200040, China.,Neurosurgery Department of Huashan Hospital, 12 Wulumuqi Zhong Road, Shanghai, 200040, China
| | - Zhengda Yu
- Glioma Surgery Division, Neurosurgery Department of Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Xiaobo Chen
- Department of Radiology and BRIC, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Islem Rekik
- Department of Radiology and BRIC, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,BASIRA Lab, CVIP Group, School of Science and Engineering, Computing, University of Dundee, Dundee, UK
| | - Qian Wang
- Med-X Research Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai, 200030, China.
| | - Junfeng Lu
- Glioma Surgery Division, Neurosurgery Department of Huashan Hospital, Fudan University, Shanghai, 200040, China. .,Shanghai Key Lab of Medical Image Computing and Computer-Assisted Intervention, Shanghai, 200040, China. .,Neurosurgery Department of Huashan Hospital, 12 Wulumuqi Zhong Road, Shanghai, 200040, China.
| | - Dinggang Shen
- Department of Radiology and BRIC, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA. .,Department of Brain and Cognitive Engineering, Korea University, Seoul, 02841, Republic of Korea.
| |
Collapse
|
15
|
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
|
16
|
Cui H, Wang X, Bian Y, Song S, Feng DD. Ischemic stroke clinical outcome prediction based on image signature selection from multimodality data. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2019; 2018:722-725. [PMID: 30440498 DOI: 10.1109/embc.2018.8512291] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Quantitative models are essential in precision medicine that can be used to predict health status and prevent disease and disability. Current radiomics models for clinical outcome prediction often depend on huge amount of image features and may include redundant information and ignore individual feature importance. In this work, we propose a prognostic discrimination ranking strategy to select the most relevant image features for image assisted clinical outcome prediction. Firstly, a redundancy and prognostic discrimination evaluation method is proposed to evaluate and rank a large number of features extracted from images. Secondly, forward sequential feature selection is performed to select the top ranked relevant features in each discriminate quantization. Finally, representative vectors are generated by the fusion of pivotal clinical parameters and selected image signatures to be fed into a classification model. The proposed model was trained and tested over 70 patient studies with six MR sequences and four clinical parameters from ISLES challenges. The evaluations using ROC curves demonstrated the improved performance over five other feature selection models where the proposed model achieved AUCs of 0.821, 0.968, 0.983, 0.896 and 1 when predicting five clinical outcome scores respectively.
Collapse
|
17
|
Prediction of IDH1 Mutation Status in Glioblastoma Using Machine Learning Technique Based on Quantitative Radiomic Data. World Neurosurg 2019; 125:e688-e696. [DOI: 10.1016/j.wneu.2019.01.157] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 01/14/2019] [Accepted: 01/17/2019] [Indexed: 12/22/2022]
|
18
|
Sarkiss CA, Germano IM. Machine Learning in Neuro-Oncology: Can Data Analysis From 5346 Patients Change Decision-Making Paradigms? World Neurosurg 2019; 124:287-294. [PMID: 30684706 DOI: 10.1016/j.wneu.2019.01.046] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 01/13/2019] [Accepted: 01/14/2019] [Indexed: 12/30/2022]
Abstract
BACKGROUND Machine learning (ML) is an application of artificial intelligence (AI) that gives computer systems the ability to learn data, without being explicitly programmed. Currently, ML has been successfully used for optical character recognition, spam filtering, and face recognition. The aim of the present study was to review the current applications of ML in the field of neuro-oncology. METHODS We conducted a systematic literature review using the PubMed and Cochrane databases using a keyword search for January 30, 2000 to March 31, 2018. The data were clustered for neuro-oncology scope of ML into 3 categories: patient outcome predictors, imaging analysis, and gene expression. RESULTS Data from 5346 patients in 29 studies were used to develop ML-based algorithms (MLBAs) in neuro-oncology. MLBAs were used to predict the outcomes for 2483 patients, with a sensitivity range of 78%-98% and specificity range of 76%-95%. In all studies, the MLBAs had greater accuracy than the conventional ones. MLBAs for image analysis showed accuracy in diagnosing low-grade versus high-grade gliomas, ranging from 80% to 93% and 90% for diagnosing high-grade glioma versus lymphoma. Seven studies used MLBAs to analyze gene expression in neuro-oncology. CONCLUSIONS MLBAs in neuro-oncology have been shown to predict patients' outcomes more accurately than conventional parameters in a retrospective analysis. If their high diagnostic accuracy in imaging analysis and detection of somatic mutations are corroborated in prospective studies, the use of tissue diagnosis or liquid biopsy might be curtailed. Finally, MLBAs are promising to help guide targeted therapy, can lead to personalized medicine, and open areas of study in the cancer cellular signaling system, not otherwise known.
Collapse
Affiliation(s)
- Christopher A Sarkiss
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, Mount Sinai Health System, New York, New York, USA
| | - Isabelle M Germano
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, Mount Sinai Health System, New York, New York, USA; Department of Economics, New York University Leonard N. Stern School of Business, New York University, New York, New York, USA.
| |
Collapse
|
19
|
Nie D, Lu J, Zhang H, Adeli E, Wang J, Yu Z, Liu L, Wang Q, Wu J, Shen D. Multi-Channel 3D Deep Feature Learning for Survival Time Prediction of Brain Tumor Patients Using Multi-Modal Neuroimages. Sci Rep 2019; 9:1103. [PMID: 30705340 PMCID: PMC6355868 DOI: 10.1038/s41598-018-37387-9] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 11/13/2018] [Indexed: 12/17/2022] Open
Abstract
High-grade gliomas are the most aggressive malignant brain tumors. Accurate pre-operative prognosis for this cohort can lead to better treatment planning. Conventional survival prediction based on clinical information is subjective and could be inaccurate. Recent radiomics studies have shown better prognosis by using carefully-engineered image features from magnetic resonance images (MRI). However, feature engineering is usually time consuming, laborious and subjective. Most importantly, the engineered features cannot effectively encode other predictive but implicit information provided by multi-modal neuroimages. We propose a two-stage learning-based method to predict the overall survival (OS) time of high-grade gliomas patient. At the first stage, we adopt deep learning, a recently dominant technique of artificial intelligence, to automatically extract implicit and high-level features from multi-modal, multi-channel preoperative MRI such that the features are competent of predicting survival time. Specifically, we utilize not only contrast-enhanced T1 MRI, but also diffusion tensor imaging (DTI) and resting-state functional MRI (rs-fMRI), for computing multiple metric maps (including various diffusivity metric maps derived from DTI, and also the frequency-specific brain fluctuation amplitude maps and local functional connectivity anisotropy-related metric maps derived from rs-fMRI) from 68 high-grade glioma patients with different survival time. We propose a multi-channel architecture of 3D convolutional neural networks (CNNs) for deep learning upon those metric maps, from which high-level predictive features are extracted for each individual patch of these maps. At the second stage, those deeply learned features along with the pivotal limited demographic and tumor-related features (such as age, tumor size and histological type) are fed into a support vector machine (SVM) to generate the final prediction result (i.e., long or short overall survival time). The experimental results demonstrate that this multi-model, multi-channel deep survival prediction framework achieves an accuracy of 90.66%, outperforming all the competing methods. This study indicates highly demanded effectiveness on prognosis of deep learning technique in neuro-oncological applications for better individualized treatment planning towards precision medicine.
Collapse
Affiliation(s)
- Dong Nie
- Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27514, USA.,Department of Radiology and BRIC, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27514, USA
| | - Junfeng Lu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200040, China.,Shanghai Key Lab of Medical Image Computing and Computer Assisted Intervention, Shanghai, 200040, China
| | - Han Zhang
- Department of Radiology and BRIC, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27514, USA
| | - Ehsan Adeli
- Department of Radiology and BRIC, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27514, USA
| | - Jun Wang
- Department of Radiology and BRIC, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27514, USA
| | - Zhengda Yu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200040, China.,Shanghai Key Lab of Medical Image Computing and Computer Assisted Intervention, Shanghai, 200040, China
| | - LuYan Liu
- Med-X Research Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Qian Wang
- Med-X Research Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China.
| | - Jinsong Wu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200040, China. .,Shanghai Key Lab of Medical Image Computing and Computer Assisted Intervention, Shanghai, 200040, China.
| | - Dinggang Shen
- Department of Radiology and BRIC, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27514, USA. .,Department of Brain and Cognitive Engineering, Korea University, Seoul, 02841, Republic of Korea.
| |
Collapse
|
20
|
Pérez-Beteta J, Martínez-González A, Pérez-García VM. A three-dimensional computational analysis of magnetic resonance images characterizes the biological aggressiveness in malignant brain tumours. J R Soc Interface 2018; 15:20180503. [PMID: 30958226 PMCID: PMC6303800 DOI: 10.1098/rsif.2018.0503] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 11/05/2018] [Indexed: 12/30/2022] Open
Abstract
Glioblastoma (GBM) is the most frequent and aggressive type of primary brain tumour. The development of image-based biomarkers from magnetic resonance images (MRIs) has been a topic of recent interest. GBMs on pre-treatment post-contrast T1-weighted (w) MRIs often appear as rim-shaped regions. In this research, we wanted to define rim-shape complexity (RSC) descriptors and study their value as indicators of the tumour's biological aggressiveness. We constructed a set of widths characterizing the rim-shaped contrast-enhancing areas in T1w MRIs, defined measures of the RSC and computed them for 311 GBM patients. Survival analysis, correlations and sensitivity studies were performed to assess the prognostic value of the measurements. All measures obtained from the histograms were found to depend on the class width to some extent. Several measures (FWHM and βR) had high prognostic value. Some histogram-independent measures were predictors of survival: maximum rim width, mean rim width and spherically averaged rim width. The later quantity allowed patients to be classified into subgroups with different rates of survival (mean difference 6.28 months, p = 0.006). In conclusion, some of the morphological quantifiers obtained from pre-treatment T1w MRIs provided information on the biological aggressiveness of GBMs. The results can be used to define prognostic measurements of clinical applicability.
Collapse
Affiliation(s)
- J. Pérez-Beteta
- Department of Mathematics, Mathematical Oncology Laboratory (MôLAB), University of Castilla-La Mancha, 13071 Ciudad Real, Spain
| | | | | |
Collapse
|
21
|
Wu G, Shi Z, Chen Y, Wang Y, Yu J, Lv X, Chen L, Ju X, Chen Z. A sparse representation-based radiomics for outcome prediction of higher grade gliomas. Med Phys 2018; 46:250-261. [PMID: 30418680 DOI: 10.1002/mp.13288] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 10/29/2018] [Accepted: 10/30/2018] [Indexed: 12/24/2022] Open
Abstract
PURPOSE Accurately predicting outcome (i.e., overall survival (OS) time) for higher grade glioma (HGG) has great clinical value and would provide optimized guidelines for treatment planning. Radiomics focuses on revealing underlying pathophysiological information in biomedical images for disease analysis and demonstrates promising prognostic clinical performance. In this paper, we propose a novel sparse representation-based radiomics framework to predict if HGG patients would have long or short OS time. METHODS First, taking advantages of the scale invariant feature transform (SIFT) feature in image characterizing, we developed a sparse representation-based method to convert a local SIFT descriptor into a global tumor feature. Next, because preserving sample structure is beneficial for feature selection, we proposed a locality preserving projection and sparse representation-combined feature selection method to select more discriminative features for tumor classification. Finally, we employed a multifeature collaborative sparse representation classification to combine the information of multimodal images to classify OS time. RESULTS Three experiments were performed on the two datasets provided by different institutions. Specifically, the proposed model was trained and independently tested on dataset 1 (135 subjects), on dataset 2 (86 subjects), and on the combination of dataset 1 and dataset 2, respectively. Experimental results demonstrated that the proposed method achieved encouraging prediction performance, exhibiting a testing accuracy of 93.33% on dataset 1 (one modality), 92.31% on dataset 2 (two modalities), and 87.93% on the combined dataset (one modality). CONCLUSIONS The sparse representation theory provides reasonable solutions to feature extraction, feature selection, and classification for radiomics. This study provides a promising tool to enhance the prediction performance of HGG patient's outcome.
Collapse
Affiliation(s)
- Guoqing Wu
- Department of Electronic Engineering, Fudan University, Shanghai, 200433, China
| | - Zhifeng Shi
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200433, China
| | - Yinsheng Chen
- Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510000, China
| | - Yuanyuan Wang
- Department of Electronic Engineering, Fudan University and Key Laboratory of Medical Imaging Computing and Computer Assisted Intervention of Shanghai, Shanghai, 200433, China
| | - Jinhua Yu
- Department of Electronic Engineering, Fudan University and Key Laboratory of Medical Imaging Computing and Computer Assisted Intervention of Shanghai, Shanghai, 200433, China
| | - Xiaofei Lv
- Department of Medical Imaging, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510000, China
| | - Liang Chen
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200433, China
| | - Xue Ju
- Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510000, China
| | - Zhongping Chen
- Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510000, China
| |
Collapse
|
22
|
Vergun S, Suhonen JI, Nair VA, Kuo J, Baskaya M, Garcia-Ramos C, Meyerand EE, Prabhakaran V. Predicting primary outcomes of brain tumor patients with advanced neuroimaging MRI measures. INTERDISCIPLINARY NEUROSURGERY : ADVANCED TECHNIQUES AND CASE MANAGEMENT 2018; 13:109-118. [PMID: 34984173 PMCID: PMC8722581 DOI: 10.1016/j.inat.2018.04.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
BACKGROUND Advanced neuroimaging measures along with clinical variables acquired during standard imaging protocols provide a rich source of information for brain tumor patient treatment and management. Machine learning analysis has had much recent success in neuroimaging applications for normal and patient populations and has potential, specifically for brain tumor patient outcome prediction. The purpose of this work was to construct, using the current patient population distribution, a high accuracy predictor for brain tumor patient outcomes of mortality and morbidity (i.e., transient and persistent language and motor deficits). The clinical value offered is a statistical tool to help guide treatment and planning as well as an investigation of the influential factors of the disease process. METHODS Resting state fMRI, diffusion tensor imaging, and task fMRI data in combination with clinical and demographic variables were used to represent the tumor patient population (n = 62; mean age = 51.2 yrs.) in a machine learning analysis in order to predict outcomes. RESULTS A support vector machine classifier with a t-test filter and recursive feature elimination predicted patient mortality (18-month interval) with 80.7% accuracy, language deficits (transient) with 74.2%, motor deficits with 71.0%, language outcomes (persistent) with 80.7% and motor outcomes with 83.9%. The most influential features of the predictors were resting fMRI connectivity, and fractional anisotropy and mean diffusivity measures in the internal capsule, brain stem and superior and inferior longitudinal fasciculi. CONCLUSIONS This study showed that advanced neuroimaging data with machine learning methods can potentially predict patient outcomes and reveal influential factors driving the predictions.
Collapse
Affiliation(s)
- Svyat Vergun
- Department of Medical Physics, University of Wisconsin-Madison, School of Medicine and Public Health, 1111 Highland Avenue, Madison, WI53792-3252, USA
| | - Josh I. Suhonen
- Department of Radiology, University of Wisconsin-Madison, School of Medicine and Public Health, 600 Highland Avenue, Wisconsin Institutes for Medical Research (WIMR), Madison, WI 53705, USA
| | - Veena A. Nair
- Department of Radiology, University of Wisconsin-Madison, School of Medicine and Public Health, 600 Highland Avenue, Wisconsin Institutes for Medical Research (WIMR), Madison, WI 53705, USA
| | - J.S. Kuo
- Department of Neurosurgery, University of Wisconsin-Madison, School of Medicine and Public Health, University of Wisconsin, Box 8660 Clinical Science Center, 600 Highland Ave, Madison, WI 53792, USA
| | - M.K. Baskaya
- Department of Neurosurgery, University of Wisconsin-Madison, School of Medicine and Public Health, University of Wisconsin, Box 8660 Clinical Science Center, 600 Highland Ave, Madison, WI 53792, USA
| | - Camille Garcia-Ramos
- Department of Medical Physics, University of Wisconsin-Madison, School of Medicine and Public Health, 1111 Highland Avenue, Madison, WI53792-3252, USA
| | - Elizabeth E. Meyerand
- Departments of Biomedical Engineering University of Wisconsin-Madison, 1550 Engineering Dr, Madison, WI 53706, USA
- Medical Physics, University of Wisconsin-Madison, 1111 Highland Ave., Suite 1129, Wisconsin Institutes for Medical Research (WIMR), Madison, WI 53705, USA
| | - Vivek Prabhakaran
- Department of Radiology, Director of Functional Neuroimaging in Radiology, University of Wisconsin Madison, School of Medicine and Public Health, 600 Highland Avenue, Madison, WI 53792-3252, USA
| |
Collapse
|
23
|
Citak-Er F, Firat Z, Kovanlikaya I, Ture U, Ozturk-Isik E. Machine-learning in grading of gliomas based on multi-parametric magnetic resonance imaging at 3T. Comput Biol Med 2018; 99:154-160. [PMID: 29933126 DOI: 10.1016/j.compbiomed.2018.06.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 06/10/2018] [Accepted: 06/11/2018] [Indexed: 10/28/2022]
Abstract
OBJECTIVE The objective of this study was to assess the contribution of multi-parametric (mp) magnetic resonance imaging (MRI) quantitative features in the machine learning-based grading of gliomas with a multi-region-of-interests approach. MATERIALS AND METHODS Forty-three patients who were newly diagnosed as having a glioma were included in this study. The patients were scanned prior to any therapy using a standard brain tumor magnetic resonance (MR) imaging protocol that included T1 and T2-weighted, diffusion-weighted, diffusion tensor, MR perfusion and MR spectroscopic imaging. Three different regions-of-interest were drawn for each subject to encompass tumor, immediate tumor periphery, and distant peritumoral edema/normal. The normalized mp-MRI features were used to build machine-learning models for differentiating low-grade gliomas (WHO grades I and II) from high grades (WHO grades III and IV). In order to assess the contribution of regional mp-MRI quantitative features to the classification models, a support vector machine-based recursive feature elimination method was applied prior to classification. RESULTS A machine-learning model based on support vector machine algorithm with linear kernel achieved an accuracy of 93.0%, a specificity of 86.7%, and a sensitivity of 96.4% for the grading of gliomas using ten-fold cross validation based on the proposed subset of the mp-MRI features. CONCLUSION In this study, machine-learning based on multiregional and multi-parametric MRI data has proven to be an important tool in grading glial tumors accurately even in this limited patient population. Future studies are needed to investigate the use of machine learning algorithms for brain tumor classification in a larger patient cohort.
Collapse
Affiliation(s)
- Fusun Citak-Er
- Department of Computer Programming, Pîrî Reis University, Istanbul, Turkey; Department of Biotechnology, Yeditepe University, Istanbul, Turkey.
| | - Zeynep Firat
- Department of Radiology, Yeditepe University Hospital, Istanbul, Turkey
| | - Ilhami Kovanlikaya
- Department of Radiology, Weill Cornell Medical College, New York, NY, USA
| | - Ugur Ture
- Department of Neurosurgery, Yeditepe University Hospital, Istanbul, Turkey
| | - Esin Ozturk-Isik
- Biomedical Engineering Institute, Boğaziçi University, Istanbul, Turkey
| |
Collapse
|
24
|
De Looze C, Beausang A, Cryan J, Loftus T, Buckley PG, Farrell M, Looby S, Reilly R, Brett F, Kearney H. Machine learning: a useful radiological adjunct in determination of a newly diagnosed glioma's grade and IDH status. J Neurooncol 2018; 139:491-499. [PMID: 29770897 DOI: 10.1007/s11060-018-2895-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 05/02/2018] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Machine learning methods have been introduced as a computer aided diagnostic tool, with applications to glioma characterisation on MRI. Such an algorithmic approach may provide a useful adjunct for a rapid and accurate diagnosis of a glioma. The aim of this study is to devise a machine learning algorithm that may be used by radiologists in routine practice to aid diagnosis of both: WHO grade and IDH mutation status in de novo gliomas. METHODS To evaluate the status quo, we interrogated the accuracy of neuroradiology reports in relation to WHO grade: grade II 96.49% (95% confidence intervals [CI] 0.88, 0.99); III 36.51% (95% CI 0.24, 0.50); IV 72.9% (95% CI 0.67, 0.78). We derived five MRI parameters from the same diagnostic brain scans, in under two minutes per case, and then supplied these data to a random forest algorithm. RESULTS Machine learning resulted in a high level of accuracy in prediction of tumour grade: grade II/III; area under the receiver operating characteristic curve (AUC) = 98%, sensitivity = 0.82, specificity = 0.94; grade II/IV; AUC = 100%, sensitivity = 1.0, specificity = 1.0; grade III/IV; AUC = 97%, sensitivity = 0.83, specificity = 0.97. Furthermore, machine learning also facilitated the discrimination of IDH status: AUC of 88%, sensitivity = 0.81, specificity = 0.77. CONCLUSIONS These data demonstrate the ability of machine learning to accurately classify diffuse gliomas by both WHO grade and IDH status from routine MRI alone-without significant image processing, which may facilitate usage as a diagnostic adjunct in clinical practice.
Collapse
Affiliation(s)
- Céline De Looze
- Trinity Centre for Bioengineering, Trinity College Dublin, Dublin, Ireland.,School of Engineering, Trinity College Dublin, Dublin, Ireland
| | - Alan Beausang
- Department of Neuropathology, Beaumont Hospital, Dublin, Ireland
| | - Jane Cryan
- Department of Neuropathology, Beaumont Hospital, Dublin, Ireland
| | - Teresa Loftus
- Department of Molecular Pathology, Beaumont Hospital, Dublin, Ireland
| | - Patrick G Buckley
- Department of Molecular Pathology, Beaumont Hospital, Dublin, Ireland.,Genomics Medicine Ireland, Dublin, Ireland
| | - Michael Farrell
- Department of Neuropathology, Beaumont Hospital, Dublin, Ireland
| | - Seamus Looby
- Department of Neuroradiology, Beaumont Hospital, Dublin, Ireland
| | - Richard Reilly
- Trinity Centre for Bioengineering, Trinity College Dublin, Dublin, Ireland.,Institute of Neurosciences, Trinity College Dublin, Dublin, Ireland.,School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Francesca Brett
- Department of Neuropathology, Beaumont Hospital, Dublin, Ireland
| | - Hugh Kearney
- Department of Neuropathology, Beaumont Hospital, Dublin, Ireland.
| |
Collapse
|
25
|
Park YW, Han K, Ahn SS, Choi YS, Chang JH, Kim SH, Kang SG, Kim EH, Lee SK. Whole-Tumor Histogram and Texture Analyses of DTI for Evaluation of IDH1-Mutation and 1p/19q-Codeletion Status in World Health Organization Grade II Gliomas. AJNR Am J Neuroradiol 2018. [PMID: 29519794 DOI: 10.3174/ajnr.a5569] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND PURPOSE Prediction of the isocitrate dehydrogenase 1 (IDH1)-mutation and 1p/19q-codeletion status of World Health Organization grade ll gliomas preoperatively may assist in predicting prognosis and planning treatment strategies. Our aim was to characterize the histogram and texture analyses of apparent diffusion coefficient and fractional anisotropy maps to determine IDH1-mutation and 1p/19q-codeletion status in World Health Organization grade II gliomas. MATERIALS AND METHODS Ninety-three patients with World Health Organization grade II gliomas with known IDH1-mutation and 1p/19q-codeletion status (18 IDH1 wild-type, 45 IDH1 mutant and no 1p/19q codeletion, 30 IDH1-mutant and 1p/19q codeleted tumors) underwent DTI. ROIs were drawn on every section of the T2-weighted images and transferred to the ADC and the fractional anisotropy maps to derive volume-based data of the entire tumor. Histogram and texture analyses were correlated with the IDH1-mutation and 1p/19q-codeletion status. The predictive powers of imaging features for IDH1 wild-type tumors and 1p/19q-codeletion status in IDH1-mutant subgroups were evaluated using the least absolute shrinkage and selection operator. RESULTS Various histogram and texture parameters differed significantly according to IDH1-mutation and 1p/19q-codeletion status. The skewness and energy of ADC, 10th and 25th percentiles, and correlation of fractional anisotropy were independent predictors of an IDH1 wild-type in the least absolute shrinkage and selection operator. The area under the receiver operating curve for the prediction model was 0.853. The skewness and cluster shade of ADC, energy, and correlation of fractional anisotropy were independent predictors of a 1p/19q codeletion in IDH1-mutant tumors in the least absolute shrinkage and selection operator. The area under the receiver operating curve was 0.807. CONCLUSIONS Whole-tumor histogram and texture features of the ADC and fractional anisotropy maps are useful for predicting the IDH1-mutation and 1p/19q-codeletion status in World Health Organization grade II gliomas.
Collapse
Affiliation(s)
- Y W Park
- From the Department of Radiology (Y.W.P.), Ewha Womans University College of Medicine, Seoul, Korea.,Departments of Radiology and Research Institute of Radiological Science (Y.W.P., K.H., S.S.A., Y.S.C., S.-K.L.)
| | - K Han
- Departments of Radiology and Research Institute of Radiological Science (Y.W.P., K.H., S.S.A., Y.S.C., S.-K.L.)
| | - S S Ahn
- Departments of Radiology and Research Institute of Radiological Science (Y.W.P., K.H., S.S.A., Y.S.C., S.-K.L.)
| | - Y S Choi
- Departments of Radiology and Research Institute of Radiological Science (Y.W.P., K.H., S.S.A., Y.S.C., S.-K.L.)
| | - J H Chang
- Neurosurgery (J.H.C., S.-G.K., E.H.K.)
| | - S H Kim
- Pathology (S.H.K.), Yonsei University College of Medicine, Seoul, Korea
| | - S-G Kang
- Neurosurgery (J.H.C., S.-G.K., E.H.K.)
| | - E H Kim
- Neurosurgery (J.H.C., S.-G.K., E.H.K.)
| | - S-K Lee
- Departments of Radiology and Research Institute of Radiological Science (Y.W.P., K.H., S.S.A., Y.S.C., S.-K.L.)
| |
Collapse
|
26
|
Abstract
OBJECTIVES Analysis of a single slice of a tumor to extract biomarkers for texture analysis may result in loss of information. We investigated correlation of fractional volumes to entire tumor volumes and introduced expanded regions of interest (ROIs) outside the visual tumor borders in glioblastoma. MATERIALS AND METHODS Retrospective slice-by-slice volumetric texture analysis on 46 brain magnetic resonance imaging subjects with histologically confirmed glioblastoma was performed. Fractional volumes were analyzed for correlation to total volume. Expanded ROIs were analyzed for significant differences to conservative ROIs. RESULTS As fractional tumor volumes increased, correlation with total volume values for mean, SD, mean of positive pixels, skewness, and kurtosis increased. Expanding ROI by 2 mm resulted in significant differences in all textural values. CONCLUSIONS Fractional volumes may provide an optimal trade-off for texture analysis in the clinical setting. All texture parameters proved significantly different with minimal expansion of the ROI, underlining the susceptibility of texture analysis to generating misrepresentative tumor information.
Collapse
|
27
|
Limkin EJ, Sun R, Dercle L, Zacharaki EI, Robert C, Reuzé S, Schernberg A, Paragios N, Deutsch E, Ferté C. Promises and challenges for the implementation of computational medical imaging (radiomics) in oncology. Ann Oncol 2018; 28:1191-1206. [PMID: 28168275 DOI: 10.1093/annonc/mdx034] [Citation(s) in RCA: 530] [Impact Index Per Article: 75.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Medical image processing and analysis (also known as Radiomics) is a rapidly growing discipline that maps digital medical images into quantitative data, with the end goal of generating imaging biomarkers as decision support tools for clinical practice. The use of imaging data from routine clinical work-up has tremendous potential in improving cancer care by heightening understanding of tumor biology and aiding in the implementation of precision medicine. As a noninvasive method of assessing the tumor and its microenvironment in their entirety, radiomics allows the evaluation and monitoring of tumor characteristics such as temporal and spatial heterogeneity. One can observe a rapid increase in the number of computational medical imaging publications-milestones that have highlighted the utility of imaging biomarkers in oncology. Nevertheless, the use of radiomics as clinical biomarkers still necessitates amelioration and standardization in order to achieve routine clinical adoption. This Review addresses the critical issues to ensure the proper development of radiomics as a biomarker and facilitate its implementation in clinical practice.
Collapse
Affiliation(s)
- E J Limkin
- Radiomics team, INSERM U1030, Gustave Roussy.,Department of Radiotherapy, Gustave Roussy, Paris-Saclay University, Villejuif
| | - R Sun
- Radiomics team, INSERM U1030, Gustave Roussy.,Department of Radiotherapy, Gustave Roussy, Paris-Saclay University, Villejuif.,Faculty of Medicine, Paris Sud University, Kremlin-Bicetre
| | - L Dercle
- Department of Nuclear Medicine and Endocrine Oncology, Gustave Roussy, Paris-Saclay University, Villejuif
| | - E I Zacharaki
- Center for Visual Computing, CentraleSupelec/Paris-Saclay University/Inria, Châtenay-Malabry
| | - C Robert
- Radiomics team, INSERM U1030, Gustave Roussy.,Department of Radiotherapy, Gustave Roussy, Paris-Saclay University, Villejuif.,Faculty of Medicine, Paris Sud University, Kremlin-Bicetre
| | - S Reuzé
- Radiomics team, INSERM U1030, Gustave Roussy.,Department of Radiotherapy, Gustave Roussy, Paris-Saclay University, Villejuif.,Faculty of Medicine, Paris Sud University, Kremlin-Bicetre
| | - A Schernberg
- Radiomics team, INSERM U1030, Gustave Roussy.,Department of Radiotherapy, Gustave Roussy, Paris-Saclay University, Villejuif.,Faculty of Medicine, Paris Sud University, Kremlin-Bicetre
| | - N Paragios
- Center for Visual Computing, CentraleSupelec/Paris-Saclay University/Inria, Châtenay-Malabry.,TheraPanacea, Paris
| | - E Deutsch
- Radiomics team, INSERM U1030, Gustave Roussy.,Department of Radiotherapy, Gustave Roussy, Paris-Saclay University, Villejuif
| | - C Ferté
- Radiomics team, INSERM U1030, Gustave Roussy.,Department of Head and Neck Oncology, Gustave Roussy, Paris-Saclay University, Villejuif, France
| |
Collapse
|
28
|
Jakola AS, Zhang YH, Skjulsvik AJ, Solheim O, Bø HK, Berntsen EM, Reinertsen I, Gulati S, Förander P, Brismar TB. Quantitative texture analysis in the prediction of IDH status in low-grade gliomas. Clin Neurol Neurosurg 2018; 164:114-120. [DOI: 10.1016/j.clineuro.2017.12.007] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 11/30/2017] [Accepted: 12/04/2017] [Indexed: 01/17/2023]
|
29
|
Wu G, Wang Y, Yu J. Overall Survival Time Prediction for High Grade Gliomas Based on Sparse Representation Framework. BRAINLESION: GLIOMA, MULTIPLE SCLEROSIS, STROKE AND TRAUMATIC BRAIN INJURIES 2018. [DOI: 10.1007/978-3-319-75238-9_7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
30
|
Sun R, Limkin E, Dercle L, Reuzé S, Zacharaki E, Chargari C, Schernberg A, Dirand A, Alexis A, Paragios N, Deutsch É, Ferté C, Robert C. Imagerie médicale computationnelle (radiomique) et potentiel en immuno-oncologie. Cancer Radiother 2017; 21:648-654. [DOI: 10.1016/j.canrad.2017.07.035] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Accepted: 07/01/2017] [Indexed: 12/12/2022]
|
31
|
Kanas VG, Zacharaki EI, Thomas GA, Zinn PO, Megalooikonomou V, Colen RR. Learning MRI-based classification models for MGMT methylation status prediction in glioblastoma. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2017; 140:249-257. [PMID: 28254081 DOI: 10.1016/j.cmpb.2016.12.018] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 12/14/2016] [Accepted: 12/29/2016] [Indexed: 06/06/2023]
Abstract
BACKGROUND AND OBJECTIVE The O6-methylguanine-DNA-methyltransferase (MGMT) promoter methylation has been shown to be associated with improved outcomes in patients with glioblastoma (GBM) and may be a predictive marker of sensitivity to chemotherapy. However, determination of the MGMT promoter methylation status requires tissue obtained via surgical resection or biopsy. The aim of this study was to assess the ability of quantitative and qualitative imaging variables in predicting MGMT methylation status noninvasively. METHODS A retrospective analysis of MR images from GBM patients was conducted. Multivariate prediction models were obtained by machine-learning methods and tested on data from The Cancer Genome Atlas (TCGA) database. RESULTS The status of MGMT promoter methylation was predicted with an accuracy of up to 73.6%. Experimental analysis showed that the edema/necrosis volume ratio, tumor/necrosis volume ratio, edema volume, and tumor location and enhancement characteristics were the most significant variables in respect to the status of MGMT promoter methylation in GBM. CONCLUSIONS The obtained results provide further evidence of an association between standard preoperative MRI variables and MGMT methylation status in GBM.
Collapse
Affiliation(s)
- Vasileios G Kanas
- Department of Electrical and Computer Engineering, University of Patras, Patras, Greece; Department of Computer Engineering and Informatics, University of Patras, Patras, Greece
| | - Evangelia I Zacharaki
- Department of Computer Engineering and Informatics, University of Patras, Patras, Greece; Center for Visual Computing (CVC), CentraleSupélec, INRIA, Université Paris-Saclay, France.
| | - Ginu A Thomas
- Department of Diagnostic Radiology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Pascal O Zinn
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX, USA
| | | | - Rivka R Colen
- Department of Diagnostic Radiology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
32
|
Zhang B, Chang K, Ramkissoon S, Tanguturi S, Bi WL, Reardon DA, Ligon KL, Alexander BM, Wen PY, Huang RY. Multimodal MRI features predict isocitrate dehydrogenase genotype in high-grade gliomas. Neuro Oncol 2017; 19:109-117. [PMID: 27353503 PMCID: PMC5193019 DOI: 10.1093/neuonc/now121] [Citation(s) in RCA: 178] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND High-grade gliomas with mutations in the isocitrate dehydrogenase (IDH) gene family confer longer overall survival relative to their IDH-wild-type counterparts. Accurate determination of the IDH genotype preoperatively may have both prognostic and diagnostic value. The current study used a machine-learning algorithm to generate a model predictive of IDH genotype in high-grade gliomas based on clinical variables and multimodal features extracted from conventional MRI. METHODS Preoperative MRIs were obtained for 120 patients with primary grades III (n = 35) and IV (n = 85) glioma in this retrospective study. IDH genotype was confirmed for grade III (32/35, 91%) and IV (22/85, 26%) tumors by immunohistochemistry, spectrometry-based mutation genotyping (OncoMap), or multiplex exome sequencing (OncoPanel). IDH1 and IDH2 mutations were mutually exclusive, and all mutated tumors were collapsed into one IDH-mutated cohort. Cases were randomly assigned to either the training (n = 90) or validation cohort (n = 30). A total of 2970 imaging features were extracted from pre- and postcontrast T1-weighted, T2-weighted, and apparent diffusion coefficient map. Using a random forest algorithm, nonredundant features were integrated with clinical data to generate a model predictive of IDH genotype. RESULTS Our model achieved accuracies of 86% (area under the curve [AUC] = 0.8830) in the training cohort and 89% (AUC = 0.9231) in the validation cohort. Features with the highest predictive value included patient age as well as parametric intensity, texture, and shape features. CONCLUSION Using a machine-learning algorithm, we achieved accurate prediction of IDH genotype in high-grade gliomas with preoperative clinical and MRI features.
Collapse
Affiliation(s)
- Biqi Zhang
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts (B.Z., K.C., R.Y.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Harvard Medical School, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts(S.R., D.A.R., K.L.L., P.Y.W.); Harvard Radiation Oncology Program, Boston, Massachusetts (S.T.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (W.L.B.); Center of Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Radiation Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (B.M.A.)
| | - Ken Chang
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts (B.Z., K.C., R.Y.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Harvard Medical School, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts(S.R., D.A.R., K.L.L., P.Y.W.); Harvard Radiation Oncology Program, Boston, Massachusetts (S.T.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (W.L.B.); Center of Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Radiation Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (B.M.A.)
| | - Shakti Ramkissoon
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts (B.Z., K.C., R.Y.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Harvard Medical School, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts(S.R., D.A.R., K.L.L., P.Y.W.); Harvard Radiation Oncology Program, Boston, Massachusetts (S.T.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (W.L.B.); Center of Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Radiation Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (B.M.A.)
| | - Shyam Tanguturi
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts (B.Z., K.C., R.Y.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Harvard Medical School, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts(S.R., D.A.R., K.L.L., P.Y.W.); Harvard Radiation Oncology Program, Boston, Massachusetts (S.T.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (W.L.B.); Center of Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Radiation Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (B.M.A.)
| | - Wenya Linda Bi
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts (B.Z., K.C., R.Y.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Harvard Medical School, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts(S.R., D.A.R., K.L.L., P.Y.W.); Harvard Radiation Oncology Program, Boston, Massachusetts (S.T.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (W.L.B.); Center of Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Radiation Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (B.M.A.)
| | - David A Reardon
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts (B.Z., K.C., R.Y.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Harvard Medical School, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts(S.R., D.A.R., K.L.L., P.Y.W.); Harvard Radiation Oncology Program, Boston, Massachusetts (S.T.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (W.L.B.); Center of Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Radiation Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (B.M.A.)
| | - Keith L Ligon
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts (B.Z., K.C., R.Y.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Harvard Medical School, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts(S.R., D.A.R., K.L.L., P.Y.W.); Harvard Radiation Oncology Program, Boston, Massachusetts (S.T.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (W.L.B.); Center of Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Radiation Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (B.M.A.)
| | - Brian M Alexander
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts (B.Z., K.C., R.Y.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Harvard Medical School, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts(S.R., D.A.R., K.L.L., P.Y.W.); Harvard Radiation Oncology Program, Boston, Massachusetts (S.T.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (W.L.B.); Center of Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Radiation Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (B.M.A.)
| | - Patrick Y Wen
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts (B.Z., K.C., R.Y.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Harvard Medical School, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts(S.R., D.A.R., K.L.L., P.Y.W.); Harvard Radiation Oncology Program, Boston, Massachusetts (S.T.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (W.L.B.); Center of Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Radiation Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (B.M.A.)
| | - Raymond Y Huang
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts (B.Z., K.C., R.Y.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Harvard Medical School, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts(S.R., D.A.R., K.L.L., P.Y.W.); Harvard Radiation Oncology Program, Boston, Massachusetts (S.T.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (W.L.B.); Center of Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Radiation Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (B.M.A.)
| |
Collapse
|
33
|
Chaddad A, Desrosiers C, Hassan L, Tanougast C. A quantitative study of shape descriptors from glioblastoma multiforme phenotypes for predicting survival outcome. Br J Radiol 2016; 89:20160575. [PMID: 27781499 DOI: 10.1259/bjr.20160575] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
OBJECTIVE Predicting the survival outcome of patients with glioblastoma multiforme (GBM) is of key importance to clinicians for selecting the optimal course of treatment. The goal of this study was to evaluate the usefulness of geometric shape features, extracted from MR images, as a potential non-invasive way to characterize GBM tumours and predict the overall survival times of patients with GBM. METHODS The data of 40 patients with GBM were obtained from the Cancer Genome Atlas and Cancer Imaging Archive. The T1 weighted post-contrast and fluid-attenuated inversion-recovery volumes of patients were co-registered and segmented into delineate regions corresponding to three GBM phenotypes: necrosis, active tumour and oedema/invasion. A set of two-dimensional shape features were then extracted slicewise from each phenotype region and combined over slices to describe the three-dimensional shape of these phenotypes. Thereafter, a Kruskal-Wallis test was employed to identify shape features with significantly different distributions across phenotypes. Moreover, a Kaplan-Meier analysis was performed to find features strongly associated with GBM survival. Finally, a multivariate analysis based on the random forest model was used for predicting the survival group of patients with GBM. RESULTS Our analysis using the Kruskal-Wallis test showed that all but one shape feature had statistically significant differences across phenotypes, with p-value < 0.05, following Holm-Bonferroni correction, justifying the analysis of GBM tumour shapes on a per-phenotype basis. Furthermore, the survival analysis based on the Kaplan-Meier estimator identified three features derived from necrotic regions (i.e. Eccentricity, Extent and Solidity) that were significantly correlated with overall survival (corrected p-value < 0.05; hazard ratios between 1.68 and 1.87). In the multivariate analysis, features from necrotic regions gave the highest accuracy in predicting the survival group of patients, with a mean area under the receiver-operating characteristic curve (AUC) of 63.85%. Combining the features of all three phenotypes increased the mean AUC to 66.99%, suggesting that shape features from different phenotypes can be used in a synergic manner to predict GBM survival. CONCLUSION Results show that shape features, in particular those extracted from necrotic regions, can be used effectively to characterize GBM tumours and predict the overall survival of patients with GBM. Advances in knowledge: Simple volumetric features have been largely used to characterize the different phenotypes of a GBM tumour (i.e. active tumour, oedema and necrosis). This study extends previous work by considering a wide range of shape features, extracted in different phenotypes, for the prediction of survival in patients with GBM.
Collapse
Affiliation(s)
- Ahmad Chaddad
- 1 Laboratory for Imagery, Vision and Artificial Intelligence, University of Québec, École de Technologie Supérieure, Montréal, QC, Canada.,2 Laboratory of Conception, Optimization and Modeling of Systems, University of Lorraine, Metz, Lorraine, France
| | - Christian Desrosiers
- 1 Laboratory for Imagery, Vision and Artificial Intelligence, University of Québec, École de Technologie Supérieure, Montréal, QC, Canada
| | - Lama Hassan
- 1 Laboratory for Imagery, Vision and Artificial Intelligence, University of Québec, École de Technologie Supérieure, Montréal, QC, Canada.,2 Laboratory of Conception, Optimization and Modeling of Systems, University of Lorraine, Metz, Lorraine, France
| | - Camel Tanougast
- 2 Laboratory of Conception, Optimization and Modeling of Systems, University of Lorraine, Metz, Lorraine, France
| |
Collapse
|
34
|
Liu L, Zhang H, Rekik I, Chen X, Wang Q, Shen D. Outcome Prediction for Patient with High-Grade Gliomas from Brain Functional and Structural Networks. MEDICAL IMAGE COMPUTING AND COMPUTER-ASSISTED INTERVENTION : MICCAI ... INTERNATIONAL CONFERENCE ON MEDICAL IMAGE COMPUTING AND COMPUTER-ASSISTED INTERVENTION 2016; 9901:26-34. [PMID: 28649677 PMCID: PMC5479332 DOI: 10.1007/978-3-319-46723-8_4] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
High-grade glioma (HGG) is a lethal cancer, which is characterized by very poor prognosis. To help optimize treatment strategy, accurate preoperative prediction of HGG patient's outcome (i.e., survival time) is of great clinical value. However, there are huge individual variability of HGG, which produces a large variation in survival time, thus making prognostic prediction more challenging. Previous brain imaging-based outcome prediction studies relied only on the imaging intensity inside or slightly around the tumor, while ignoring any information that is located far away from the lesion (i.e., the "normal appearing" brain tissue). Notably, in addition to altering MR image intensity, we hypothesize that the HGG growth and its mass effect also change both structural (can be modeled by diffusion tensor imaging (DTI)) and functional brain connectivities (estimated by functional magnetic resonance imaging (rs-fMRI)). Therefore, integrating connectomics information in outcome prediction could improve prediction accuracy. To this end, we unprecedentedly devise a machine learning-based HGG prediction framework that can effectively extract valuable features from complex human brain connectome using network analysis tools, followed by a novel multi-stage feature selection strategy to single out good features while reducing feature redundancy. Ultimately, we use support vector machine (SVM) to classify HGG outcome as either bad (survival time ≤ 650 days) or good (survival time >650 days). Our method achieved 75 % prediction accuracy. We also found that functional and structural networks provide complementary information for the outcome prediction, thus leading to increased prediction accuracy compared with the baseline method, which only uses the basic clinical information (63.2 %).
Collapse
Affiliation(s)
- Luyan Liu
- Med-X Research Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Han Zhang
- Department of Radiology and BRIC, University of North Carolina at Chapel Hill, Chapel Hill, USA
| | - Islem Rekik
- Department of Radiology and BRIC, University of North Carolina at Chapel Hill, Chapel Hill, USA
| | - Xiaobo Chen
- Department of Radiology and BRIC, University of North Carolina at Chapel Hill, Chapel Hill, USA
| | - Qian Wang
- Med-X Research Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Dinggang Shen
- Department of Radiology and BRIC, University of North Carolina at Chapel Hill, Chapel Hill, USA
| |
Collapse
|
35
|
Nie D, Zhang H, Adeli E, Liu L, Shen D. 3D Deep Learning for Multi-modal Imaging-Guided Survival Time Prediction of Brain Tumor Patients. MEDICAL IMAGE COMPUTING AND COMPUTER-ASSISTED INTERVENTION : MICCAI ... INTERNATIONAL CONFERENCE ON MEDICAL IMAGE COMPUTING AND COMPUTER-ASSISTED INTERVENTION 2016; 9901:212-220. [PMID: 28149967 DOI: 10.1007/978-3-319-46723-8_25] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
High-grade glioma is the most aggressive and severe brain tumor that leads to death of almost 50% patients in 1-2 years. Thus, accurate prognosis for glioma patients would provide essential guidelines for their treatment planning. Conventional survival prediction generally utilizes clinical information and limited handcrafted features from magnetic resonance images (MRI), which is often time consuming, laborious and subjective. In this paper, we propose using deep learning frameworks to automatically extract features from multi-modal preoperative brain images (i.e., T1 MRI, fMRI and DTI) of high-grade glioma patients. Specifically, we adopt 3D convolutional neural networks (CNNs) and also propose a new network architecture for using multi-channel data and learning supervised features. Along with the pivotal clinical features, we finally train a support vector machine to predict if the patient has a long or short overall survival (OS) time. Experimental results demonstrate that our methods can achieve an accuracy as high as 89.9% We also find that the learned features from fMRI and DTI play more important roles in accurately predicting the OS time, which provides valuable insights into functional neuro-oncological applications.
Collapse
Affiliation(s)
- Dong Nie
- Department of Radiology and BRIC, University of North Carolina at Chapel Hill, Chapel Hill, USA; Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, USA
| | - Han Zhang
- Department of Radiology and BRIC, University of North Carolina at Chapel Hill, Chapel Hill, USA
| | - Ehsan Adeli
- Department of Radiology and BRIC, University of North Carolina at Chapel Hill, Chapel Hill, USA
| | - Luyan Liu
- Department of Radiology and BRIC, University of North Carolina at Chapel Hill, Chapel Hill, USA
| | - Dinggang Shen
- Department of Radiology and BRIC, University of North Carolina at Chapel Hill, Chapel Hill, USA
| |
Collapse
|
36
|
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
|
37
|
Burth S, Kickingereder P, Eidel O, Tichy D, Bonekamp D, Weberling L, Wick A, Löw S, Hertenstein A, Nowosielski M, Schlemmer HP, Wick W, Bendszus M, Radbruch A. Clinical parameters outweigh diffusion- and perfusion-derived MRI parameters in predicting survival in newly diagnosed glioblastoma. Neuro Oncol 2016; 18:1673-1679. [PMID: 27298312 DOI: 10.1093/neuonc/now122] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 05/03/2016] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The purpose of this study was to determine the relevance of clinical data, apparent diffusion coefficient (ADC), and relative cerebral blood volume (rCBV) from dynamic susceptibility contrast (DSC) perfusion and the volume transfer constant (ktrans) from dynamic contrast-enhanced (DCE) perfusion for predicting overall survival (OS) and progression-free survival (PFS) in newly diagnosed treatment-naïve glioblastoma patients. METHODS Preoperative MR scans including standardized contrast-enhanced T1 (cT1), T2 - fluid-attenuated inversion recovery (FLAIR), ADC, DSC, and DCE of 125 patients with subsequent histopathologically confirmed glioblastoma were performed on a 3 Tesla MRI scanner. ADC, DSC, and DCE parameters were analyzed in semiautomatically segmented tumor volumes on contrast-enhanced (CE) cT1 and hyperintense signal changes on T2 FLAIR (ED). Univariate and multivariable Cox regression analyses including age, sex, extent of resection (EOR), and KPS were performed to assess the influence of each parameter on OS and PFS. RESULTS Univariate Cox regression analysis demonstrated a significant association of age, KPS, and EOR with PFS and age, KPS, EOR, lower ADC, and higher rCBV with OS. Multivariable analysis showed independent significance of male sex, KPS, EOR, and increased rCBVCE for PFS, and age, sex, KPS, and EOR for OS. CONCLUSIONS MRI parameters help to predict OS in a univariate Cox regression analysis, and increased rCBVCE is associated with shorter PFS in the multivariable model. In summary, however, our findings suggest that the relevance of MRI parameters is outperformed by clinical parameters in a multivariable analysis, which limits their prognostic value for survival prediction at the time of initial diagnosis.
Collapse
Affiliation(s)
- Sina Burth
- Department of Neuroradiology, University of Heidelberg Medical Center, Heidelberg, Germany (S.B., P.K., O.E., D.B., L.W., M.B., A.R.); Division of Bioststatistics, German Cancer Research Center, Heidelberg, Germany (D.T.); Department of Neurology, University of Heidelberg Medical Center, Heidelberg, Germany (A.W., S.L., A.H., W.W.); Department of Neurology, Innsbruck Medical University, Innsbruck, Austria (M.N.); Department of Radiology, German Cancer Research Center, Heidelberg, Germany (H.S.)
| | - Philipp Kickingereder
- Department of Neuroradiology, University of Heidelberg Medical Center, Heidelberg, Germany (S.B., P.K., O.E., D.B., L.W., M.B., A.R.); Division of Bioststatistics, German Cancer Research Center, Heidelberg, Germany (D.T.); Department of Neurology, University of Heidelberg Medical Center, Heidelberg, Germany (A.W., S.L., A.H., W.W.); Department of Neurology, Innsbruck Medical University, Innsbruck, Austria (M.N.); Department of Radiology, German Cancer Research Center, Heidelberg, Germany (H.S.)
| | - Oliver Eidel
- Department of Neuroradiology, University of Heidelberg Medical Center, Heidelberg, Germany (S.B., P.K., O.E., D.B., L.W., M.B., A.R.); Division of Bioststatistics, German Cancer Research Center, Heidelberg, Germany (D.T.); Department of Neurology, University of Heidelberg Medical Center, Heidelberg, Germany (A.W., S.L., A.H., W.W.); Department of Neurology, Innsbruck Medical University, Innsbruck, Austria (M.N.); Department of Radiology, German Cancer Research Center, Heidelberg, Germany (H.S.)
| | - Diana Tichy
- Department of Neuroradiology, University of Heidelberg Medical Center, Heidelberg, Germany (S.B., P.K., O.E., D.B., L.W., M.B., A.R.); Division of Bioststatistics, German Cancer Research Center, Heidelberg, Germany (D.T.); Department of Neurology, University of Heidelberg Medical Center, Heidelberg, Germany (A.W., S.L., A.H., W.W.); Department of Neurology, Innsbruck Medical University, Innsbruck, Austria (M.N.); Department of Radiology, German Cancer Research Center, Heidelberg, Germany (H.S.)
| | - David Bonekamp
- Department of Neuroradiology, University of Heidelberg Medical Center, Heidelberg, Germany (S.B., P.K., O.E., D.B., L.W., M.B., A.R.); Division of Bioststatistics, German Cancer Research Center, Heidelberg, Germany (D.T.); Department of Neurology, University of Heidelberg Medical Center, Heidelberg, Germany (A.W., S.L., A.H., W.W.); Department of Neurology, Innsbruck Medical University, Innsbruck, Austria (M.N.); Department of Radiology, German Cancer Research Center, Heidelberg, Germany (H.S.)
| | - Lukas Weberling
- Department of Neuroradiology, University of Heidelberg Medical Center, Heidelberg, Germany (S.B., P.K., O.E., D.B., L.W., M.B., A.R.); Division of Bioststatistics, German Cancer Research Center, Heidelberg, Germany (D.T.); Department of Neurology, University of Heidelberg Medical Center, Heidelberg, Germany (A.W., S.L., A.H., W.W.); Department of Neurology, Innsbruck Medical University, Innsbruck, Austria (M.N.); Department of Radiology, German Cancer Research Center, Heidelberg, Germany (H.S.)
| | - Antje Wick
- Department of Neuroradiology, University of Heidelberg Medical Center, Heidelberg, Germany (S.B., P.K., O.E., D.B., L.W., M.B., A.R.); Division of Bioststatistics, German Cancer Research Center, Heidelberg, Germany (D.T.); Department of Neurology, University of Heidelberg Medical Center, Heidelberg, Germany (A.W., S.L., A.H., W.W.); Department of Neurology, Innsbruck Medical University, Innsbruck, Austria (M.N.); Department of Radiology, German Cancer Research Center, Heidelberg, Germany (H.S.)
| | - Sarah Löw
- Department of Neuroradiology, University of Heidelberg Medical Center, Heidelberg, Germany (S.B., P.K., O.E., D.B., L.W., M.B., A.R.); Division of Bioststatistics, German Cancer Research Center, Heidelberg, Germany (D.T.); Department of Neurology, University of Heidelberg Medical Center, Heidelberg, Germany (A.W., S.L., A.H., W.W.); Department of Neurology, Innsbruck Medical University, Innsbruck, Austria (M.N.); Department of Radiology, German Cancer Research Center, Heidelberg, Germany (H.S.)
| | - Anne Hertenstein
- Department of Neuroradiology, University of Heidelberg Medical Center, Heidelberg, Germany (S.B., P.K., O.E., D.B., L.W., M.B., A.R.); Division of Bioststatistics, German Cancer Research Center, Heidelberg, Germany (D.T.); Department of Neurology, University of Heidelberg Medical Center, Heidelberg, Germany (A.W., S.L., A.H., W.W.); Department of Neurology, Innsbruck Medical University, Innsbruck, Austria (M.N.); Department of Radiology, German Cancer Research Center, Heidelberg, Germany (H.S.)
| | - Martha Nowosielski
- Department of Neuroradiology, University of Heidelberg Medical Center, Heidelberg, Germany (S.B., P.K., O.E., D.B., L.W., M.B., A.R.); Division of Bioststatistics, German Cancer Research Center, Heidelberg, Germany (D.T.); Department of Neurology, University of Heidelberg Medical Center, Heidelberg, Germany (A.W., S.L., A.H., W.W.); Department of Neurology, Innsbruck Medical University, Innsbruck, Austria (M.N.); Department of Radiology, German Cancer Research Center, Heidelberg, Germany (H.S.)
| | - Heinz-Peter Schlemmer
- Department of Neuroradiology, University of Heidelberg Medical Center, Heidelberg, Germany (S.B., P.K., O.E., D.B., L.W., M.B., A.R.); Division of Bioststatistics, German Cancer Research Center, Heidelberg, Germany (D.T.); Department of Neurology, University of Heidelberg Medical Center, Heidelberg, Germany (A.W., S.L., A.H., W.W.); Department of Neurology, Innsbruck Medical University, Innsbruck, Austria (M.N.); Department of Radiology, German Cancer Research Center, Heidelberg, Germany (H.S.)
| | - Wolfgang Wick
- Department of Neuroradiology, University of Heidelberg Medical Center, Heidelberg, Germany (S.B., P.K., O.E., D.B., L.W., M.B., A.R.); Division of Bioststatistics, German Cancer Research Center, Heidelberg, Germany (D.T.); Department of Neurology, University of Heidelberg Medical Center, Heidelberg, Germany (A.W., S.L., A.H., W.W.); Department of Neurology, Innsbruck Medical University, Innsbruck, Austria (M.N.); Department of Radiology, German Cancer Research Center, Heidelberg, Germany (H.S.)
| | - Martin Bendszus
- Department of Neuroradiology, University of Heidelberg Medical Center, Heidelberg, Germany (S.B., P.K., O.E., D.B., L.W., M.B., A.R.); Division of Bioststatistics, German Cancer Research Center, Heidelberg, Germany (D.T.); Department of Neurology, University of Heidelberg Medical Center, Heidelberg, Germany (A.W., S.L., A.H., W.W.); Department of Neurology, Innsbruck Medical University, Innsbruck, Austria (M.N.); Department of Radiology, German Cancer Research Center, Heidelberg, Germany (H.S.)
| | - Alexander Radbruch
- Department of Neuroradiology, University of Heidelberg Medical Center, Heidelberg, Germany (S.B., P.K., O.E., D.B., L.W., M.B., A.R.); Division of Bioststatistics, German Cancer Research Center, Heidelberg, Germany (D.T.); Department of Neurology, University of Heidelberg Medical Center, Heidelberg, Germany (A.W., S.L., A.H., W.W.); Department of Neurology, Innsbruck Medical University, Innsbruck, Austria (M.N.); Department of Radiology, German Cancer Research Center, Heidelberg, Germany (H.S.)
| |
Collapse
|
38
|
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
|
39
|
Federau C, Cerny M, Roux M, Mosimann PJ, Maeder P, Meuli R, Wintermark M. IVIM perfusion fraction is prognostic for survival in brain glioma. Clin Neuroradiol 2016; 27:485-492. [DOI: 10.1007/s00062-016-0510-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 03/01/2016] [Indexed: 11/30/2022]
|
40
|
Rao A, Manyam G, Rao G, Jain R. Integrative Analysis of mRNA, microRNA, and Protein Correlates of Relative Cerebral Blood Volume Values in GBM Reveals the Role for Modulators of Angiogenesis and Tumor Proliferation. Cancer Inform 2016; 15:29-33. [PMID: 27053917 PMCID: PMC4814129 DOI: 10.4137/cin.s33014] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 11/29/2016] [Accepted: 12/07/2015] [Indexed: 12/12/2022] Open
Abstract
Dynamic susceptibility contrast-enhanced magnetic resonance imaging is routinely used to provide hemodynamic assessment of brain tumors as a diagnostic as well as a prognostic tool. Recently, it was shown that the relative cerebral blood volume (rCBV), obtained from the contrast-enhancing as well as -nonenhancing portion of glioblastoma (GBM), is strongly associated with overall survival. In this study, we aim to characterize the genomic correlates (microRNA, messenger RNA, and protein) of this vascular parameter. This study aims to provide a comprehensive radiogenomic and radioproteomic characterization of the hemodynamic phenotype of GBM using publicly available imaging and genomic data from the Cancer Genome Atlas GBM cohort. Based on this analysis, we identified pathways associated with angiogenesis and tumor proliferation underlying this hemodynamic parameter in GBM.
Collapse
Affiliation(s)
- Arvind Rao
- Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ganiraju Manyam
- Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ganesh Rao
- Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rajan Jain
- Department of Radiology, NY University School of Medicine, New York, NY, USA
| |
Collapse
|
41
|
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
|
42
|
Hoffermann M, Bruckmann L, Mahdy Ali K, Asslaber M, Payer F, von Campe G. Treatment results and outcome in elderly patients with glioblastoma multiforme – A retrospective single institution analysis. Clin Neurol Neurosurg 2015; 128:60-9. [DOI: 10.1016/j.clineuro.2014.11.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 10/14/2014] [Accepted: 11/09/2014] [Indexed: 10/24/2022]
|
43
|
Emblem KE, Pinho MC, Zöllner FG, Due-Tonnessen P, Hald JK, Schad LR, Meling TR, Rapalino O, Bjornerud A. A generic support vector machine model for preoperative glioma survival associations. Radiology 2014; 275:228-34. [PMID: 25486589 DOI: 10.1148/radiol.14140770] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
PURPOSE To develop a generic support vector machine (SVM) model by using magnetic resonance (MR) imaging-based blood volume distribution data for preoperative glioma survival associations and to prospectively evaluate the diagnostic effectiveness of this model in autonomous patient data. MATERIALS AND METHODS Institutional and regional medical ethics committees approved the study, and all patients signed a consent form. Two hundred thirty-five preoperative adult patients from two institutions with a subsequent histologically confirmed diagnosis of glioma after surgery were included retrospectively. An SVM learning technique was applied to MR imaging-based whole-tumor relative cerebral blood volume (rCBV) histograms. SVM models with the highest diagnostic accuracy for 6-month and 1-, 2-, and 3-year survival associations were trained on 101 patients from the first institution. With Cox survival analysis, the diagnostic effectiveness of the SVM models was tested on independent data from 134 patients at the second institution. RESULTS were adjusted for known survival predictors, including patient age, tumor size, neurologic status, and postsurgery treatment, and were compared with survival associations from an expert reader. RESULTS Compared with total qualitative assessment by an expert reader, the whole-tumor rCBV-based SVM model was the strongest parameter associated with 6-month and 1-, 2-, and 3-year survival in the independent patient data (area under the receiver operating characteristic curve, 0.794-0.851; hazard ratio, 5.4-21.2). DISCUSSION Machine learning by means of SVM in combination with whole-tumor rCBV histogram analysis can be used to identify early patient survival in aggressive gliomas. The SVM model returned higher diagnostic accuracy values than an expert reader, and the model appears to be insensitive to patient, observer, and institutional variations.
Collapse
Affiliation(s)
- Kyrre E Emblem
- From the Intervention Centre (K.E.E., A.B.), Department of Radiology (P.D.T., J.K.H.), and Department of Neurosurgery (T.R.M.), Oslo University Hospital, N-0027 Sognsvannsveien 20, 0372 Oslo, Norway; Department of Radiology and Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, Mass (K.E.E., M.C.P., O.R.); Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Tex (M.C.P.); Department of Computer Assisted Clinical Medicine, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany (F.G.Z., L.R.S.); and Department of Physics, University of Oslo, Oslo, Norway (A.B.)
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Keunen O, Taxt T, Grüner R, Lund-Johansen M, Tonn JC, Pavlin T, Bjerkvig R, Niclou SP, Thorsen F. Multimodal imaging of gliomas in the context of evolving cellular and molecular therapies. Adv Drug Deliv Rev 2014; 76:98-115. [PMID: 25078721 DOI: 10.1016/j.addr.2014.07.010] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 07/14/2014] [Accepted: 07/22/2014] [Indexed: 01/18/2023]
Abstract
The vast majority of malignant gliomas relapse after surgery and standard radio-chemotherapy. Novel molecular and cellular therapies are thus being developed, targeting specific aspects of tumor growth. While histopathology remains the gold standard for tumor classification, neuroimaging has over the years taken a central role in the diagnosis and treatment follow up of brain tumors. It is used to detect and localize lesions, define the target area for biopsies, plan surgical and radiation interventions and assess tumor progression and treatment outcome. In recent years the application of novel drugs including anti-angiogenic agents that affect the tumor vasculature, has drastically modulated the outcome of brain tumor imaging. To properly evaluate the effects of emerging experimental therapies and successfully support treatment decisions, neuroimaging will have to evolve. Multi-modal imaging systems with existing and new contrast agents, molecular tracers, technological advances and advanced data analysis can all contribute to the establishment of disease relevant biomarkers that will improve disease management and patient care. In this review, we address the challenges of glioma imaging in the context of novel molecular and cellular therapies, and take a prospective look at emerging experimental and pre-clinical imaging techniques that bear the promise of meeting these challenges.
Collapse
|
45
|
Computer-extracted MR imaging features are associated with survival in glioblastoma patients. J Neurooncol 2014; 120:483-8. [PMID: 25151504 DOI: 10.1007/s11060-014-1580-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Accepted: 08/05/2014] [Indexed: 10/24/2022]
Abstract
Automatic survival prognosis in glioblastoma (GBM) could result in improved treatment planning for the patient. The purpose of this research is to investigate the association of survival in GBM patients with tumor features in pre-operative magnetic resonance (MR) images assessed using a fully automatic computer algorithm. MR imaging data for 68 patients from two US institutions were used in this study. The images were obtained from the Cancer Imaging Archive. A fully automatic computer vision algorithm was applied to segment the images and extract eight imaging features from the MRI studies. The features included tumor side, proportion of enhancing tumor, proportion of necrosis, T1/FLAIR ratio, major axis length, minor axis length, tumor volume, and thickness of enhancing margin. We constructed a multivariate Cox proportional hazards regression model and used a likelihood ratio test to establish whether the imaging features are prognostic of survival. We also evaluated the individual prognostic value of each feature through multivariate analysis using the multivariate Cox model and univariate analysis using univariate Cox models for each feature. We found that the automatically extracted imaging features were predictive of survival (p = 0.031). Multivariate analysis of individual features showed that two individual features were predictive of survival: proportion of enhancing tumor (p = 0.013), and major axis length (p = 0.026). Univariate analysis indicated the same two features as significant (p = 0.021, and p = 0.017 respectively). We conclude that computer-extracted MR imaging features can be used for survival prognosis in GBM patients.
Collapse
|
46
|
Hilario A, Sepulveda JM, Perez-Nuñez A, Salvador E, Millan JM, Hernandez-Lain A, Rodriguez-Gonzalez V, Lagares A, Ramos A. A prognostic model based on preoperative MRI predicts overall survival in patients with diffuse gliomas. AJNR Am J Neuroradiol 2014; 35:1096-102. [PMID: 24457819 PMCID: PMC7965146 DOI: 10.3174/ajnr.a3837] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Accepted: 11/10/2013] [Indexed: 11/07/2022]
Abstract
BACKGROUND AND PURPOSE Diffuse gliomas are classified as grades II-IV on the basis of histologic features, with prognosis determined mainly by clinical factors and histologic grade supported by molecular markers. Our aim was to evaluate, in patients with diffuse gliomas, the relationship of relative CBV and ADC values to overall survival. In addition, we also propose a prognostic model based on preoperative MR imaging findings that predicts survival independent of histopathology. MATERIALS AND METHODS We conducted a retrospective analysis of the preoperative diffusion and perfusion MR imaging in 126 histologically confirmed diffuse gliomas. Median relative CBV and ADC values were selected for quantitative analysis. Survival univariate analysis was made by constructing survival curves by using the Kaplan-Meier method and comparing subgroups by log-rank probability tests. A Cox regression model was made for multivariate analysis. RESULTS The study included 126 diffuse gliomas (median follow-up of 14.5 months). ADC and relative CBV values had a significant influence on overall survival. Median overall survival for patients with ADC < 0.799 × 10(-3) mm(2)/s was <1 year. Multivariate analysis revealed that patient age, relative CBV, and ADC values were associated with survival independent of pathology. The preoperative model provides greater ability to predict survival than that obtained by histologic grade alone. CONCLUSIONS ADC values had a better correlation with overall survival than relative CBV values. A preoperative prognostic model based on patient age, relative CBV, and ADC values predicted overall survival of patients with diffuse gliomas independent of pathology. This preoperative model provides a more accurate predictor of survival than histologic grade alone.
Collapse
Affiliation(s)
- A Hilario
- From the Departments of Radiology (A.H., A.R., E.S., J.M.M.)
| | | | - A Perez-Nuñez
- Neurosurgery (A.P.-N., A.L.), Hospital 12 de Octubre, Madrid, Spain
| | - E Salvador
- From the Departments of Radiology (A.H., A.R., E.S., J.M.M.)
| | - J M Millan
- From the Departments of Radiology (A.H., A.R., E.S., J.M.M.)
| | | | | | - A Lagares
- Neurosurgery (A.P.-N., A.L.), Hospital 12 de Octubre, Madrid, Spain
| | - A Ramos
- From the Departments of Radiology (A.H., A.R., E.S., J.M.M.)
| |
Collapse
|
47
|
Svolos P, Kousi E, Kapsalaki E, Theodorou K, Fezoulidis I, Kappas C, Tsougos I. The role of diffusion and perfusion weighted imaging in the differential diagnosis of cerebral tumors: a review and future perspectives. Cancer Imaging 2014; 14:20. [PMID: 25609475 PMCID: PMC4331825 DOI: 10.1186/1470-7330-14-20] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 03/20/2014] [Indexed: 12/31/2022] Open
Abstract
The role of conventional Magnetic Resonance Imaging (MRI) in the detection of cerebral tumors has been well established. However its excellent soft tissue visualization and variety of imaging sequences are in many cases non-specific for the assessment of brain tumor grading. Hence, advanced MRI techniques, like Diffusion-Weighted Imaging (DWI), Diffusion Tensor Imaging (DTI) and Dynamic-Susceptibility Contrast Imaging (DSCI), which are based on different contrast principles, have been used in the clinical routine to improve diagnostic accuracy. The variety of quantitative information derived from these techniques provides significant structural and functional information in a cellular level, highlighting aspects of the underlying brain pathophysiology. The present work, reviews physical principles and recent results obtained using DWI/DTI and DSCI, in tumor characterization and grading of the most common cerebral neoplasms, and discusses how the available MR quantitative data can be utilized through advanced methods of analysis, in order to optimize clinical decision making.
Collapse
|
48
|
Zhang J, Barboriak DP, Hobbs H, Mazurowski MA. A fully automatic extraction of magnetic resonance image features in glioblastoma patients. Med Phys 2014; 41:042301. [DOI: 10.1118/1.4866218] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
|
49
|
Jensen RL, Mumert ML, Gillespie DL, Kinney AY, Schabel MC, Salzman KL. Preoperative dynamic contrast-enhanced MRI correlates with molecular markers of hypoxia and vascularity in specific areas of intratumoral microenvironment and is predictive of patient outcome. Neuro Oncol 2013; 16:280-91. [PMID: 24305704 DOI: 10.1093/neuonc/not148] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Measures of tumor vascularity and hypoxia have been correlated with glioma grade and outcome. Dynamic contrast-enhanced (DCE) MRI can noninvasively map tumor blood flow, vascularity, and permeability. In this prospective observational cohort pilot study, preoperative imaging was correlated with molecular markers of hypoxia, vascularity, proliferation, and progression-free and overall patient survival. METHODS Pharmacokinetic modeling methods were used to generate maps of tumor blood flow, extraction fraction, permeability-surface area product, transfer constant, washout rate, interstitial volume, blood volume, capillary transit time, and capillary heterogeneity from preoperative DCE-MRI data in human glioma patients. Tissue was obtained from areas of peritumoral edema, active tumor, hypoxic penumbra, and necrotic core and evaluated for vascularity, proliferation, and expression of hypoxia-regulated molecules. DCE-MRI parameter values were correlated with hypoxia-regulated protein expression at tissue sample sites. RESULTS Patient survival correlated with DCE parameters in 2 cases: capillary heterogeneity in active tumor and interstitial volume in areas of peritumoral edema. Statistically significant correlations were observed between several DCE parameters and tissue markers. In addition, MIB-1 index was predictive of overall survival (P = .044) and correlated with vascular endothelial growth factor expression in hypoxic penumbra (r = 0.7933, P = .0071) and peritumoral edema (r = 0.4546). Increased microvessel density correlated with worse patient outcome (P = .026). CONCLUSIONS Our findings suggest that DCE-MRI may facilitate noninvasive preoperative predictions of areas of tumor with increased hypoxia and proliferation. Both imaging and hypoxia biomarkers are predictive of patient outcome. This has the potential to allow unprecedented prognostic decisions and to guide therapies to specific tumor areas.
Collapse
Affiliation(s)
- Randy L Jensen
- Corresponding author: Randy L. Jensen, MD, PhD, Huntsman Cancer Institute and Departments of Neurosurgery, Radiation Oncology, Oncological Sciences, Clinical Neuroscience Center, University of Utah, 175 North Medical Drive, Salt Lake City, Utah 84132.
| | | | | | | | | | | |
Collapse
|
50
|
Emblem KE, Due-Tonnessen P, Hald JK, Bjornerud A, Pinho MC, Scheie D, Schad LR, Meling TR, Zoellner FG. Machine learning in preoperative glioma MRI: Survival associations by perfusion-based support vector machine outperforms traditional MRI. J Magn Reson Imaging 2013; 40:47-54. [DOI: 10.1002/jmri.24390] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Accepted: 07/12/2013] [Indexed: 11/12/2022] Open
Affiliation(s)
- Kyrre E. Emblem
- Department of Radiology and Athinoula A. Martinos Center for Biomedical Imaging; Massachusetts General Hospital and Harvard Medical School; Boston Massachusetts USA
- Intervention Centre; Rikshospitalet; Oslo University Hospital; Oslo Norway
| | | | - John K. Hald
- Department of Radiology; Rikshospitalet; Oslo University Hospital; Oslo Norway
| | - Atle Bjornerud
- Intervention Centre; Rikshospitalet; Oslo University Hospital; Oslo Norway
- Department of Physics; University of Oslo; Oslo Norway
| | - Marco C. Pinho
- Department of Radiology and Athinoula A. Martinos Center for Biomedical Imaging; Massachusetts General Hospital and Harvard Medical School; Boston Massachusetts USA
- Department of Radiology; University of Texas Southwestern Medical Center; Dallas Texas USA
| | - David Scheie
- Department of Pathology; Rikshospitalet; Oslo University Hospital; Oslo Norway
| | - Lothar R. Schad
- Computer Assisted Clinical Medicine; Medical Faculty Mannheim; Heidelberg University; Heidelberg Germany
| | - Torstein R. Meling
- Department of Neurosurgery; Rikshospitalet; Oslo University Hospital; Oslo Norway
| | - Frank G. Zoellner
- Computer Assisted Clinical Medicine; Medical Faculty Mannheim; Heidelberg University; Heidelberg Germany
| |
Collapse
|