1
|
Leone A, Di Napoli V, Fochi NP, Di Perna G, Spetzger U, Filimonova E, Angileri F, Carbone F, Colamaria A. Virtual Biopsy for the Prediction of MGMT Promoter Methylation in Gliomas: A Comprehensive Review of Radiomics and Deep Learning Approaches Applied to MRI. Diagnostics (Basel) 2025; 15:251. [PMID: 39941181 PMCID: PMC11816478 DOI: 10.3390/diagnostics15030251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 01/18/2025] [Accepted: 01/20/2025] [Indexed: 02/16/2025] Open
Abstract
Background/Objectives: The methylation status of the O6-methylguanine-DNA methyltransferase (MGMT) promoter in gliomas has emerged as a critical biomarker for prognosis and treatment response. Conventional methods for assessing MGMT promoter methylation, such as methylation-specific PCR, are invasive and require tissue sampling. Methods: A comprehensive literature search was performed in compliance with the updated PRISMA 2020 guidelines within electronic databases MEDLINE/PubMed, Scopus, and IEEE Xplore. Search terms, including "MGMT", "methylation", "glioma", "glioblastoma", "machine learning", "deep learning", and "radiomics", were adopted in various MeSH combinations. Original studies in the English, Italian, German, and French languages were considered for inclusion. Results: This review analyzed 34 studies conducted in the last six years, focusing on assessing MGMT methylation status using radiomics (RD), deep learning (DL), or combined approaches. These studies utilized radiological data from the public (e.g., BraTS, TCGA) and private institutional datasets. Sixteen studies focused exclusively on glioblastoma (GBM), while others included low- and high-grade gliomas. Twenty-seven studies reported diagnostic accuracy, with fourteen achieving values above 80%. The combined use of DL and RD generally resulted in higher accuracy, sensitivity, and specificity, although some studies reported lower minimum accuracy compared to studies using a single model. Conclusions: The integration of RD and DL offers a powerful, non-invasive tool for precisely recognizing MGMT promoter methylation status in gliomas, paving the way for enhanced personalized medicine in neuro-oncology. The heterogeneity of study populations, data sources, and methodologies reflected the complexity of the pipeline and machine learning algorithms, which may require general standardization to be implemented in clinical practice.
Collapse
Affiliation(s)
- Augusto Leone
- Department of Neurosurgery, Karlsruher Neurozentrum, Städtisches Klinikum Karlsruhe, 76133 Karlsruhe, Germany; (A.L.); (U.S.); (F.C.)
- Faculty of Human Medicine, Charité Universitätsmedizin, 10117 Berlin, Germany
| | - Veronica Di Napoli
- Department of Neurosurgery, University of Turin, 10124 Turin, Italy; (V.D.N.); (N.P.F.)
| | - Nicola Pio Fochi
- Department of Neurosurgery, University of Turin, 10124 Turin, Italy; (V.D.N.); (N.P.F.)
| | - Giuseppe Di Perna
- Division of Neurosurgery, “Policlinico Riuniti”, 71122 Foggia, Italy;
| | - Uwe Spetzger
- Department of Neurosurgery, Karlsruher Neurozentrum, Städtisches Klinikum Karlsruhe, 76133 Karlsruhe, Germany; (A.L.); (U.S.); (F.C.)
| | - Elena Filimonova
- Department of Neuroradiology, Federal Neurosurgical Center, 630048 Novosibirsk, Russia;
| | - Flavio Angileri
- Department of Neurosurgery, University of Messina, 98122 Messina, Italy;
| | - Francesco Carbone
- Department of Neurosurgery, Karlsruher Neurozentrum, Städtisches Klinikum Karlsruhe, 76133 Karlsruhe, Germany; (A.L.); (U.S.); (F.C.)
- Division of Neurosurgery, “Policlinico Riuniti”, 71122 Foggia, Italy;
| | - Antonio Colamaria
- Division of Neurosurgery, “Policlinico Riuniti”, 71122 Foggia, Italy;
| |
Collapse
|
2
|
Singh G, Singh A, Bae J, Manjila S, Spektor V, Prasanna P, Lignelli A. -New frontiers in domain-inspired radiomics and radiogenomics: increasing role of molecular diagnostics in CNS tumor classification and grading following WHO CNS-5 updates. Cancer Imaging 2024; 24:133. [PMID: 39375809 PMCID: PMC11460168 DOI: 10.1186/s40644-024-00769-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 08/31/2024] [Indexed: 10/09/2024] Open
Abstract
Gliomas and Glioblastomas represent a significant portion of central nervous system (CNS) tumors associated with high mortality rates and variable prognosis. In 2021, the World Health Organization (WHO) updated its Glioma classification criteria, most notably incorporating molecular markers including CDKN2A/B homozygous deletion, TERT promoter mutation, EGFR amplification, + 7/-10 chromosome copy number changes, and others into the grading and classification of adult and pediatric Gliomas. The inclusion of these markers and the corresponding introduction of new Glioma subtypes has allowed for more specific tailoring of clinical interventions and has inspired a new wave of Radiogenomic studies seeking to leverage medical imaging information to explore the diagnostic and prognostic implications of these new biomarkers. Radiomics, deep learning, and combined approaches have enabled the development of powerful computational tools for MRI analysis correlating imaging characteristics with various molecular biomarkers integrated into the updated WHO CNS-5 guidelines. Recent studies have leveraged these methods to accurately classify Gliomas in accordance with these updated molecular-based criteria based solely on non-invasive MRI, demonstrating the great promise of Radiogenomic tools. In this review, we explore the relative benefits and drawbacks of these computational frameworks and highlight the technical and clinical innovations presented by recent studies in the landscape of fast evolving molecular-based Glioma subtyping. Furthermore, the potential benefits and challenges of incorporating these tools into routine radiological workflows, aiming to enhance patient care and optimize clinical outcomes in the evolving field of CNS tumor management, have been highlighted.
Collapse
Affiliation(s)
- Gagandeep Singh
- Neuroradiology Division, Columbia University Irving Medical Center, New York, NY, USA.
| | - Annie Singh
- Atal Bihari Vajpayee Institute of Medical Sciences, New Delhi, India
| | - Joseph Bae
- Department of Biomedical Informatics, Stony Brook University, Stony Brook, USA
| | - Sunil Manjila
- Department of Neurological Surgery, Garden City Hospital, Garden City, MI, USA
| | - Vadim Spektor
- Neuroradiology Division, Columbia University Irving Medical Center, New York, NY, USA
| | - Prateek Prasanna
- Department of Biomedical Informatics, Stony Brook University, Stony Brook, USA
| | - Angela Lignelli
- Neuroradiology Division, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
3
|
Tariq R. Predicting response to chemotherapy in brain tumor patients based on MRI features. Clin Neurol Neurosurg 2024; 244:108409. [PMID: 38959786 DOI: 10.1016/j.clineuro.2024.108409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 06/27/2024] [Indexed: 07/05/2024]
Abstract
Chemotherapy in brain tumors is tailored based on tumor type, grade, and molecular markers, which are crucial for predicting responses and survival outcomes. This review summarizes the role of chemotherapy in gliomas, glioneuronal and neuronal tumors, ependymomas, choroid plexus tumors, medulloblastomas, and meningiomas, discussing standard treatment protocols and recent developments in targeted therapies.Furthermore, the studies reporting the integration of MRI-based radiomics and deep learning models for predicting treatment outcomes are reviewed. Advances in MRI-based radiomics and deep learning models have significantly enhanced the prediction of chemotherapeutic benefits, survival prediction following chemotherapy, and differentiating tumor progression with psuedoprogression. These non-invasive techniques offer valuable insights into tumor characteristics and treatment responses, facilitating personalized therapeutic strategies. Further research is warranted to refine these models and expand their applicability across different brain tumor types.
Collapse
Affiliation(s)
- Rabeet Tariq
- Department of Neurosurgery, Section of Surgery, Aga Khan University Hospital, Karachi, Pakistan.
| |
Collapse
|
4
|
Huang YR, Fan HQ, Kuang YY, Wang P, Lu S. The Relationship Between the Molecular Phenotypes of Brain Gliomas and the Imaging Features and Sensitivity of Radiotherapy and Chemotherapy. Clin Oncol (R Coll Radiol) 2024; 36:541-551. [PMID: 38821723 DOI: 10.1016/j.clon.2024.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 02/28/2024] [Accepted: 05/10/2024] [Indexed: 06/02/2024]
Abstract
Gliomas are the most common primary malignant tumors of the brain, accounting for about 80% of all central nervous system malignancies. With the development of molecular biology, the molecular phenotypes of gliomas have been shown to be closely related to the process of diagnosis and treatment. The molecular phenotype of glioma also plays an important role in guiding treatment plans and evaluating treatment effects and prognosis. However, due to the heterogeneity of the tumors and the trauma associated with the surgical removal of tumor tissue, the application of molecular phenotyping in glioma is limited. With the development of imaging technology, functional magnetic resonance imaging (MRI) can provide structural and function information about tumors in a noninvasive and radiation-free manner. MRI is very important for the diagnosis of intracranial lesions. In recent years, with the development of the technology for tumor molecular diagnosis and imaging, the use of molecular phenotype information and imaging procedures to evaluate the treatment outcome of tumors has become a hot topic. By reviewing the related literature on glioma treatment and molecular typing that has been published in the past 20 years, and referring to the latest 2020 NCCN treatment guidelines, summarizing the imaging characteristic and sensitivity of radiotherapy and chemotherapy of different molecular phenotypes of glioma. In this article, we briefly review the imaging characteristics of different molecular phenotypes in gliomas and their relationship with radiosensitivity and chemosensitivity of gliomas.
Collapse
Affiliation(s)
- Y-R Huang
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - H-Q Fan
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Y-Y Kuang
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - P Wang
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - S Lu
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China.
| |
Collapse
|
5
|
Doniselli FM, Pascuzzo R, Mazzi F, Padelli F, Moscatelli M, Akinci D'Antonoli T, Cuocolo R, Aquino D, Cuccarini V, Sconfienza LM. Quality assessment of the MRI-radiomics studies for MGMT promoter methylation prediction in glioma: a systematic review and meta-analysis. Eur Radiol 2024; 34:5802-5815. [PMID: 38308012 PMCID: PMC11364578 DOI: 10.1007/s00330-024-10594-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 12/04/2023] [Accepted: 12/31/2023] [Indexed: 02/04/2024]
Abstract
OBJECTIVES To evaluate the methodological quality and diagnostic accuracy of MRI-based radiomic studies predicting O6-methylguanine-DNA methyltransferase (MGMT) promoter methylation status in gliomas. METHODS PubMed Medline, EMBASE, and Web of Science were searched to identify MRI-based radiomic studies on MGMT methylation in gliomas published until December 31, 2022. Three raters evaluated the study methodological quality with Radiomics Quality Score (RQS, 16 components) and Transparent Reporting of a Multivariable Prediction Model for Individual Prognosis Or Diagnosis (TRIPOD, 22 items) scales. Risk of bias and applicability concerns were assessed with QUADAS-2 tool. A meta-analysis was performed to estimate the pooled area under the curve (AUC) and to assess inter-study heterogeneity. RESULTS We included 26 studies, published from 2016. The median RQS total score was 8 out of 36 (22%, range 8-44%). Thirteen studies performed external validation. All studies reported AUC or accuracy, but only 4 (15%) performed calibration and decision curve analysis. No studies performed phantom analysis, cost-effectiveness analysis, and prospective validation. The overall TRIPOD adherence score was between 50% and 70% in 16 studies and below 50% in 10 studies. The pooled AUC was 0.78 (95% CI, 0.73-0.83, I2 = 94.1%) with a high inter-study heterogeneity. Studies with external validation and including only WHO-grade IV gliomas had significantly lower AUC values (0.65; 95% CI, 0.57-0.73, p < 0.01). CONCLUSIONS Study RQS and adherence to TRIPOD guidelines was generally low. Radiomic prediction of MGMT methylation status showed great heterogeneity of results and lower performances in grade IV gliomas, which hinders its current implementation in clinical practice. CLINICAL RELEVANCE STATEMENT MGMT promoter methylation status appears to be variably correlated with MRI radiomic features; radiomic models are not sufficiently robust to be integrated into clinical practice to accurately predict MGMT promoter methylation status in patients with glioma before surgery. KEY POINTS • Adherence to the indications of TRIPOD guidelines was generally low, as was RQS total score. • MGMT promoter methylation status prediction with MRI radiomic features provided heterogeneous diagnostic accuracy results across studies. • Studies that included grade IV glioma only and performed external validation had significantly lower diagnostic accuracy than others.
Collapse
Affiliation(s)
- Fabio M Doniselli
- Neuroradiology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Giovanni Celoria 11, 20133, Milan, Italy
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, Via Luigi Mangiagalli 31, 20133, Milan, Italy
| | - Riccardo Pascuzzo
- Neuroradiology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Giovanni Celoria 11, 20133, Milan, Italy.
| | - Federica Mazzi
- Neuroradiology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Giovanni Celoria 11, 20133, Milan, Italy
| | - Francesco Padelli
- Neuroradiology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Giovanni Celoria 11, 20133, Milan, Italy
| | - Marco Moscatelli
- Neuroradiology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Giovanni Celoria 11, 20133, Milan, Italy
| | - Tugba Akinci D'Antonoli
- Institute of Radiology and Nuclear Medicine, Cantonal Hospital Baselland, Rheinstrasse 26, 4410, Liestal, Switzerland
| | - Renato Cuocolo
- Department of Medicine, Surgery, and Dentistry, University of Salerno, Via Salvador Allende 43, Baronissi, 84081, Salerno, Italy
| | - Domenico Aquino
- Neuroradiology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Giovanni Celoria 11, 20133, Milan, Italy
| | - Valeria Cuccarini
- Neuroradiology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Giovanni Celoria 11, 20133, Milan, Italy
| | - Luca Maria Sconfienza
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, Via Luigi Mangiagalli 31, 20133, Milan, Italy
- IRCCS Ospedale Galeazzi-Sant'Ambrogio, Via Cristina Belgioioso 173, 20157, Milan, Italy
| |
Collapse
|
6
|
Doddamani R, Chandra PS. Radiogenomics In Neuro-Oncology: A Noninvasive Way of Understanding Tumor Biology. Neurol India 2024; 72:698-699. [PMID: 39216019 DOI: 10.4103/neurol-india.neurol-india_88_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Affiliation(s)
- Ramesh Doddamani
- Department of Neurosurgery, All India Institute of Medical Sciences, New Delhi, India
| | | |
Collapse
|
7
|
Liu C, Wang J, Shen J, Chen X, Ji N, Yue S. Accurate and rapid molecular subgrouping of high-grade glioma via deep learning-assisted label-free fiber-optic Raman spectroscopy. PNAS NEXUS 2024; 3:pgae208. [PMID: 38860145 PMCID: PMC11164103 DOI: 10.1093/pnasnexus/pgae208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 05/17/2024] [Indexed: 06/12/2024]
Abstract
Molecular genetics is highly related with prognosis of high-grade glioma. Accordingly, the latest WHO guideline recommends that molecular subgroups of the genes, including IDH, 1p/19q, MGMT, TERT, EGFR, Chromosome 7/10, CDKN2A/B, need to be detected to better classify glioma and guide surgery and treatment. Unfortunately, there is no preoperative or intraoperative technology available for accurate and comprehensive molecular subgrouping of glioma. Here, we develop a deep learning-assisted fiber-optic Raman diagnostic platform for accurate and rapid molecular subgrouping of high-grade glioma. Specifically, a total of 2,354 fingerprint Raman spectra was obtained from 743 tissue sites (astrocytoma: 151; oligodendroglioma: 150; glioblastoma (GBM): 442) of 44 high-grade glioma patients. The convolutional neural networks (ResNet) model was then established and optimized for molecular subgrouping. The mean area under receiver operating characteristic curves (AUC) for identifying the molecular subgroups of high-grade glioma reached 0.904, with mean sensitivity of 83.3%, mean specificity of 85.0%, mean accuracy of 83.3%, and mean time expense of 10.6 s. The diagnosis performance using ResNet model was shown to be superior to PCA-SVM and UMAP models, suggesting that high dimensional information from Raman spectra would be helpful. In addition, for the molecular subgroups of GBM, the mean AUC reached 0.932, with mean sensitivity of 87.8%, mean specificity of 83.6%, and mean accuracy of 84.1%. Furthermore, according to saliency maps, the specific Raman features corresponding to tumor-associated biomolecules (e.g. nucleic acid, tyrosine, tryptophan, cholesteryl ester, fatty acid, and collagen) were found to contribute to the accurate molecular subgrouping. Collectively, this study opens up new opportunities for accurate and rapid molecular subgrouping of high-grade glioma, which would assist optimal surgical resection and instant post-operative decision-making.
Collapse
Affiliation(s)
- Chang Liu
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Institute of Medical Photonics, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Xueyuan Road 37, Beijing 100191, China
| | - Jiejun Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, South Fourth Ring West Road 119, Beijing 100050, China
| | - Jianghao Shen
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Institute of Medical Photonics, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Xueyuan Road 37, Beijing 100191, China
| | - Xun Chen
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Institute of Medical Photonics, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Xueyuan Road 37, Beijing 100191, China
- School of Engineering Medicine, Beihang University, Xueyuan Road 37, Beijing 100191, China
| | - Nan Ji
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, South Fourth Ring West Road 119, Beijing 100050, China
| | - Shuhua Yue
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Institute of Medical Photonics, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Xueyuan Road 37, Beijing 100191, China
| |
Collapse
|
8
|
Zheng F, Zhang L, Chen H, Zang Y, Chen X, Li Y. Radiomics for predicting MGMT status in cerebral glioblastoma: comparison of different MRI sequences. JOURNAL OF RADIATION RESEARCH 2024; 65:350-359. [PMID: 38650477 PMCID: PMC11115443 DOI: 10.1093/jrr/rrae007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/12/2023] [Indexed: 04/25/2024]
Abstract
Using radiomics to predict O6-methylguanine-DNA methyltransferase promoter methylation status in patients with newly diagnosed glioblastoma and compare the performances of different MRI sequences. Preoperative MRI scans from 215 patients were included in this retrospective study. After image preprocessing and feature extraction, two kinds of machine-learning models were established and compared for their performances. One kind was established using all MRI sequences (T1-weighted image, T2-weighted image, contrast enhancement, fluid-attenuated inversion recovery, DWI_b_high, DWI_b_low and apparent diffusion coefficient), and the other kind was based on single MRI sequence as listed above. For the machine-learning model based on all sequences, a total of seven radiomic features were selected with the Maximum Relevance and Minimum Redundancy algorithm. The predictive accuracy was 0.993 and 0.750 in the training and validation sets, respectively, and the area under curves were 1.000 and 0.754 in the two sets, respectively. For the machine-learning model based on single sequence, the numbers of selected features were 8, 10, 10, 13, 9, 7 and 6 for T1-weighted image, T2-weighted image, contrast enhancement, fluid-attenuated inversion recovery, DWI_b_high, DWI_b_low and apparent diffusion coefficient, respectively, with predictive accuracies of 0.797-1.000 and 0.583-0.694 in the training and validation sets, respectively, and the area under curves of 0.874-1.000 and 0.538-0.697 in the two sets, respectively. Specifically, T1-weighted image-based model performed best, while contrast enhancement-based model performed worst in the independent validation set. The machine-learning models based on seven different single MRI sequences performed differently in predicting O6-methylguanine-DNA methyltransferase status in glioblastoma, while the machine-learning model based on the combination of all sequences performed best.
Collapse
Affiliation(s)
- Fei Zheng
- Department of Radiology, Capital Medical University, Beijing Tiantan Hospital, No. 119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, P. R. China
- Department of Radiology, Peking University People’s Hospital, No. 11 Xizhimen South Street, Xicheng District, Beijing, 100044, P. R. China
| | - Lingling Zhang
- Department of Radiology, Capital Medical University, Beijing Tiantan Hospital, No. 119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, P. R. China
| | - Hongyan Chen
- Department of Radiology, Capital Medical University, Beijing Tiantan Hospital, No. 119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, P. R. China
| | - Yuying Zang
- Department of Radiology, Capital Medical University, Beijing Tiantan Hospital, No. 119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, P. R. China
| | - Xuzhu Chen
- Department of Radiology, Capital Medical University, Beijing Tiantan Hospital, No. 119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, P. R. China
| | - Yiming Li
- Department of Neurosurgery, Capital Medical University, Beijing Tiantan Hospital, No. 119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, P. R. China
| |
Collapse
|
9
|
Samartha MVS, Dubey NK, Jena B, Maheswar G, Lo WC, Saxena S. AI-driven estimation of O6 methylguanine-DNA-methyltransferase (MGMT) promoter methylation in glioblastoma patients: a systematic review with bias analysis. J Cancer Res Clin Oncol 2024; 150:57. [PMID: 38291266 PMCID: PMC10827977 DOI: 10.1007/s00432-023-05566-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 11/27/2023] [Indexed: 02/01/2024]
Abstract
BACKGROUND Accurate and non-invasive estimation of MGMT promoter methylation status in glioblastoma (GBM) patients is of paramount clinical importance, as it is a predictive biomarker associated with improved overall survival (OS). In response to the clinical need, recent studies have focused on the development of non-invasive artificial intelligence (AI)-based methods for MGMT estimation. In this systematic review, we not only delve into the technical aspects of these AI-driven MGMT estimation methods but also emphasize their profound clinical implications. Specifically, we explore the potential impact of accurate non-invasive MGMT estimation on GBM patient care and treatment decisions. METHODS Employing a PRISMA search strategy, we identified 33 relevant studies from reputable databases, including PubMed, ScienceDirect, Google Scholar, and IEEE Explore. These studies were comprehensively assessed using 21 diverse attributes, encompassing factors such as types of imaging modalities, machine learning (ML) methods, and cohort sizes, with clear rationales for attribute scoring. Subsequently, we ranked these studies and established a cutoff value to categorize them into low-bias and high-bias groups. RESULTS By analyzing the 'cumulative plot of mean score' and the 'frequency plot curve' of the studies, we determined a cutoff value of 6.00. A higher mean score indicated a lower risk of bias, with studies scoring above the cutoff mark categorized as low-bias (73%), while 27% fell into the high-bias category. CONCLUSION Our findings underscore the immense potential of AI-based machine learning (ML) and deep learning (DL) methods in non-invasively determining MGMT promoter methylation status. Importantly, the clinical significance of these AI-driven advancements lies in their capacity to transform GBM patient care by providing accurate and timely information for treatment decisions. However, the translation of these technical advancements into clinical practice presents challenges, including the need for large multi-institutional cohorts and the integration of diverse data types. Addressing these challenges will be critical in realizing the full potential of AI in improving the reliability and accessibility of MGMT estimation while lowering the risk of bias in clinical decision-making.
Collapse
Affiliation(s)
- Mullapudi Venkata Sai Samartha
- Department of Computer Science & Engineering, International Institute of Information Technology, Bhubaneswar, 751003, India
| | - Navneet Kumar Dubey
- Victory Biotechnology Co., Ltd., Taipei, 114757, Taiwan
- Executive Programme in Healthcare Management, Indian Institute of Management, Lucknow, 226013, India
| | - Biswajit Jena
- Institute of Technical Education and Research, SOA Deemed to be University, Bhubaneswar, 751030, India
| | - Gorantla Maheswar
- Department of Computer Science & Engineering, International Institute of Information Technology, Bhubaneswar, 751003, India
| | - Wen-Cheng Lo
- Division of Neurosurgery, Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan.
- Department of Neurosurgery, Taipei Medical University Hospital, Taipei, 11031, Taiwan.
- Taipei Neuroscience Institute, Taipei Medical University, Taipei, 11031, Taiwan.
| | - Sanjay Saxena
- Department of Computer Science & Engineering, International Institute of Information Technology, Bhubaneswar, 751003, India.
| |
Collapse
|
10
|
Xu H, Lv W, Zhang H, Yuan Q, Wang Q, Wu Y, Lu L. Multimodality radiomics analysis based on [ 18F]FDG PET/CT imaging and multisequence MRI: application to nasopharyngeal carcinoma prognosis. Eur Radiol 2023; 33:6677-6688. [PMID: 37060444 DOI: 10.1007/s00330-023-09606-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 01/02/2023] [Accepted: 02/13/2023] [Indexed: 04/16/2023]
Abstract
OBJECTIVES To determine whether radiomics models developed from 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG) PET/CT combined with multisequence MRI could contribute to predicting the progression-free survival (PFS) of nasopharyngeal carcinoma (NPC) patients. METHODS One hundred thirty-two NPC patients who underwent both PET/CT and MRI scanning were retrospectively enrolled (88 vs. 44 for training vs. testing). For each modality/sequence (i.e., PET, CT, T1, T1C, and T2), 1906 radiomics features were extracted from the primary tumor volume. Univariate Cox model and correlation analysis were used for feature selection. A multivariate Cox model was used to establish radiomics signature. Prognostic performances of 5 individual modality models and 12 multimodality models (3 integrations × 4 fusion strategies) were assessed by the concordance index (C-index) and log-rank test. A clinical-radiomics nomogram was built to explore the clinical utilities of radiomics signature, which was evaluated by discrimination, calibration curve, and decision curve analysis (DCA). RESULTS The radiomics signatures of individual modalities showed limited prognostic efficacy with a C-index of 0.539-0.664 in the testing cohort. Different fusion strategies exhibited a slight difference in predictive performance. The PET/CT and MRI integrated model achieved the best performance with a C-index of 0.745 (95% CI, 0.619-0.865) in the testing cohort (log-rank test, p < 0.05). Clinical-radiomics nomogram further improved the prognosis, which also showed satisfactory discrimination, calibration, and net benefit. CONCLUSIONS Multimodality radiomics analysis by combining PET/CT with multisequence MRI could potentially improve the efficacy of PFS prediction for NPC patients. KEY POINTS • Individual modality radiomics models showed limited performance in prognosis evaluation for NPC patients. • Combined PET, CT and multisequence MRI radiomics signature could improve the prognostic efficacy. • Multilevel fusion strategies exhibit comparable performance but feature-level fusion deserves more attention.
Collapse
Affiliation(s)
- Hui Xu
- School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, Guangdong, China
- Guangdong Provincial Key Laboratory of Medial Image Processing, Southern Medical University, Guangzhou, 510515, Guangdong, China
- Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, Guangzhou, 510515, Guangdong, China
- Pazhou Lab, Guangzhou, 510330, China
| | - Wenbing Lv
- School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, Guangdong, China
- Guangdong Provincial Key Laboratory of Medial Image Processing, Southern Medical University, Guangzhou, 510515, Guangdong, China
- Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, Guangzhou, 510515, Guangdong, China
- Pazhou Lab, Guangzhou, 510330, China
| | - Hao Zhang
- Department of Medical Imaging, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Qingyu Yuan
- Nanfang PET Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Quanshi Wang
- Nanfang PET Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Yuankui Wu
- Department of Medical Imaging, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| | - Lijun Lu
- School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, Guangdong, China.
- Guangdong Provincial Key Laboratory of Medial Image Processing, Southern Medical University, Guangzhou, 510515, Guangdong, China.
- Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, Guangzhou, 510515, Guangdong, China.
- Pazhou Lab, Guangzhou, 510330, China.
| |
Collapse
|
11
|
Faghani S, Khosravi B, Moassefi M, Conte GM, Erickson BJ. A Comparison of Three Different Deep Learning-Based Models to Predict the MGMT Promoter Methylation Status in Glioblastoma Using Brain MRI. J Digit Imaging 2023; 36:837-846. [PMID: 36604366 PMCID: PMC10287882 DOI: 10.1007/s10278-022-00757-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 12/07/2022] [Accepted: 12/08/2022] [Indexed: 01/06/2023] Open
Abstract
Glioblastoma (GBM) is the most common primary malignant brain tumor in adults. The standard treatment for GBM consists of surgical resection followed by concurrent chemoradiotherapy and adjuvant temozolomide. O-6-methylguanine-DNA methyltransferase (MGMT) promoter methylation status is an important prognostic biomarker that predicts the response to temozolomide and guides treatment decisions. At present, the only reliable way to determine MGMT promoter methylation status is through the analysis of tumor tissues. Considering the complications of the tissue-based methods, an imaging-based approach is preferred. This study aimed to compare three different deep learning-based approaches for predicting MGMT promoter methylation status. We obtained 576 T2WI with their corresponding tumor masks, and MGMT promoter methylation status from, The Brain Tumor Segmentation (BraTS) 2021 datasets. We developed three different models: voxel-wise, slice-wise, and whole-brain. For voxel-wise classification, methylated and unmethylated MGMT tumor masks were made into 1 and 2 with 0 background, respectively. We converted each T2WI into 32 × 32 × 32 patches. We trained a 3D-Vnet model for tumor segmentation. After inference, we constructed the whole brain volume based on the patch's coordinates. The final prediction of MGMT methylation status was made by majority voting between the predicted voxel values of the biggest connected component. For slice-wise classification, we trained an object detection model for tumor detection and MGMT methylation status prediction, then for final prediction, we used majority voting. For the whole-brain approach, we trained a 3D Densenet121 for prediction. Whole-brain, slice-wise, and voxel-wise, accuracy was 65.42% (SD 3.97%), 61.37% (SD 1.48%), and 56.84% (SD 4.38%), respectively.
Collapse
Affiliation(s)
- Shahriar Faghani
- Radiology Informatics Lab, Department of Radiology, Mayo Clinic, S.W, 200 1St Street, Rochester, MN, 55905, USA
| | - Bardia Khosravi
- Radiology Informatics Lab, Department of Radiology, Mayo Clinic, S.W, 200 1St Street, Rochester, MN, 55905, USA
| | - Mana Moassefi
- Radiology Informatics Lab, Department of Radiology, Mayo Clinic, S.W, 200 1St Street, Rochester, MN, 55905, USA
| | - Gian Marco Conte
- Radiology Informatics Lab, Department of Radiology, Mayo Clinic, S.W, 200 1St Street, Rochester, MN, 55905, USA
| | - Bradley J Erickson
- Radiology Informatics Lab, Department of Radiology, Mayo Clinic, S.W, 200 1St Street, Rochester, MN, 55905, USA.
| |
Collapse
|
12
|
Tao R, Yu X, Lu J, Wang Y, Lu W, Zhang Z, Li H, Zhou J. A deep learning nomogram of continuous glucose monitoring data for the risk prediction of diabetic retinopathy in type 2 diabetes. Phys Eng Sci Med 2023; 46:813-825. [PMID: 37041318 DOI: 10.1007/s13246-023-01254-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 03/27/2023] [Indexed: 04/13/2023]
Abstract
Continuous glucose monitoring (CGM) data analysis will provide a new perspective to analyze factors related to diabetic retinopathy (DR). However, the problem of visualizing CGM data and automatically predicting the incidence of DR from CGM is still controversial. Here, we explored the feasibility of using CGM profiles to predict DR in type 2 diabetes (T2D) by deep learning approach. This study fused deep learning with a regularized nomogram to construct a novel deep learning nomogram from CGM profiles to identify patients at high risk of DR. Specifically, a deep learning network was employed to mine the nonlinear relationship between CGM profiles and DR. Moreover, a novel nomogram combining CGM deep factors with basic information was established to score the patients' DR risk. This dataset consists of 788 patients belonging to two cohorts: 494 in the training cohort and 294 in the testing cohort. The area under the curve (AUC) values of our deep learning nomogram were 0.82 and 0.80 in the training cohort and testing cohort, respectively. By incorporating basic clinical factors, the deep learning nomogram achieved an AUC of 0.86 in the training cohort and 0.85 in the testing cohort. The calibration plot and decision curve showed that the deep learning nomogram had the potential for clinical application. This analysis method of CGM profiles can be extended to other diabetic complications by further investigation.
Collapse
Affiliation(s)
- Rui Tao
- College of Information Science and Engineering, Northeastern University, Shenyang, Liaoning, China
| | - Xia Yu
- College of Information Science and Engineering, Northeastern University, Shenyang, Liaoning, China
| | - Jingyi Lu
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Clinical Center for Diabetes, 600 Yishan Road, Shanghai, 200233, China
| | - Yaxin Wang
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Clinical Center for Diabetes, 600 Yishan Road, Shanghai, 200233, China
| | - Wei Lu
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Clinical Center for Diabetes, 600 Yishan Road, Shanghai, 200233, China
| | - Zhanhu Zhang
- College of Information Science and Engineering, Northeastern University, Shenyang, Liaoning, China
| | - Hongru Li
- College of Information Science and Engineering, Northeastern University, Shenyang, Liaoning, China
| | - Jian Zhou
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Clinical Center for Diabetes, 600 Yishan Road, Shanghai, 200233, China.
| |
Collapse
|
13
|
Wang G, Li L, Wang J, Zang J, Chen J, Xiao Y, Fan X, Zhu L, Kung HF, Zhu Z. Head-to-head comparison of [ 68Ga]Ga-P16-093 and 2-[ 18F]FDG PET/CT in patients with clear cell renal cell carcinoma: a pilot study. Eur J Nucl Med Mol Imaging 2023; 50:1499-1509. [PMID: 36600099 DOI: 10.1007/s00259-022-06101-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 12/24/2022] [Indexed: 01/06/2023]
Abstract
PURPOSE This pilot study was prospectively designed to evaluate and compare the diagnostic value of PET/CT using a PSMA-specific tracer [68Ga]Ga-P16-093 and a glucose metabolism probe 2-[18F]FDG in clear cell renal cell carcinoma (ccRCC) patients. METHODS Forty-two pathologically confirmed ccRCC patients were included. Within 1 week, each patient underwent [68Ga]Ga-P16-093 and 2-[18F]FDG PET/CT. In addition to visual analysis of tumor number, the standardized uptake value (SUV) was measured for semiquantitative comparison and correlation analysis. RESULTS For primary ccRCC patients, [68Ga]Ga-P16-093 PET/CT demonstrated a significantly higher detection rate (19/22 vs. 13/22, P = 0.031) and higher tumor uptake (15.7 ± 9.0 vs. 5.1 ± 3.4, P < 0.001) than 2-[18F]FDG PET/CT. In addition, the SUVmax of the primary tumor on [68Ga]Ga-P16-093 and 2-[18F]FDG PET/CT was significantly correlated with pT stage (for [68Ga]Ga-P16-093, r = 0.550, P = 0.008; for 2-[18F]FDG, r = 0.514, P = 0.014) and WHO/ISUP grade (for [68Ga]Ga-P16-093, r = 0.566, P = 0.006; for 2-[18F]FDG, r = 0.492, P = 0.020), respectively. For metastatic ccRCC patients, [68Ga]Ga-P16-093 PET/CT also demonstrated a better detection rate (21/22 vs. 14/22, P = 0.008) and higher tumor uptake (11.0 ± 6.4 vs. 4.4 ± 2.7, P < 0.001) than 2-[18F]FDG PET/CT. The SUVmax on [68Ga]Ga-P16-093 PET/CT had a significant association with PSMA expression in primary ccRCC (r = 0.776, P < 0.001) and metastatic ccRCC (r = 0.626, P = 0.029). CONCLUSIONS [68Ga]Ga-P16-093 PET/CT demonstrates significantly better tumor detectability than 2-[18F]FDG PET/CT for ccRCC patients. TRIAL REGISTRATION 68Ga-P16-093 and 18F-FDG PET/CT Imaging in the Same Group of Clear Cell Renal Cell Carcinoma Patients (NCT05432947, Registered 27 June 2021, retrospectively registered) URL OF REGISTRY: https://clinicaltrials.gov/ct2/show/NCT05432947 .
Collapse
Affiliation(s)
- Guochang Wang
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100730, China
| | - Linlin Li
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100730, China
| | - Jiarou Wang
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100730, China
| | - Jie Zang
- Department of Nuclear Medicine, Fujian Provincial Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - Jingci Chen
- Department of Pathology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Yu Xiao
- Department of Pathology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Xinrong Fan
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100730, China.
| | - Lin Zhu
- College of Chemistry, Key Laboratory of Radiopharmaceuticals, Ministry of Education, Beijing Normal University, Beijing, 100875, China.
| | - Hank F Kung
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| | - Zhaohui Zhu
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
14
|
Prediction of O-6-methylguanine-DNA methyltransferase and overall survival of the patients suffering from glioblastoma using MRI-based hybrid radiomics signatures in machine and deep learning framework. Neural Comput Appl 2023. [DOI: 10.1007/s00521-023-08405-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2023]
|
15
|
Qureshi SA, Hussain L, Ibrar U, Alabdulkreem E, Nour MK, Alqahtani MS, Nafie FM, Mohamed A, Mohammed GP, Duong TQ. Radiogenomic classification for MGMT promoter methylation status using multi-omics fused feature space for least invasive diagnosis through mpMRI scans. Sci Rep 2023; 13:3291. [PMID: 36841898 PMCID: PMC9961309 DOI: 10.1038/s41598-023-30309-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 02/21/2023] [Indexed: 02/27/2023] Open
Abstract
Accurate radiogenomic classification of brain tumors is important to improve the standard of diagnosis, prognosis, and treatment planning for patients with glioblastoma. In this study, we propose a novel two-stage MGMT Promoter Methylation Prediction (MGMT-PMP) system that extracts latent features fused with radiomic features predicting the genetic subtype of glioblastoma. A novel fine-tuned deep learning architecture, namely Deep Learning Radiomic Feature Extraction (DLRFE) module, is proposed for latent feature extraction that fuses the quantitative knowledge to the spatial distribution and the size of tumorous structure through radiomic features: (GLCM, HOG, and LBP). The application of the novice rejection algorithm has been found significantly effective in selecting and isolating the negative training instances out of the original dataset. The fused feature vectors are then used for training and testing by k-NN and SVM classifiers. The 2021 RSNA Brain Tumor challenge dataset (BraTS-2021) consists of four structural mpMRIs, viz. fluid-attenuated inversion-recovery, T1-weighted, T1-weighted contrast enhancement, and T2-weighted. We evaluated the classification performance, for the very first time in published form, in terms of measures like accuracy, F1-score, and Matthews correlation coefficient. The Jackknife tenfold cross-validation was used for training and testing BraTS-2021 dataset validation. The highest classification performance is (96.84 ± 0.09)%, (96.08 ± 0.10)%, and (97.44 ± 0.14)% as accuracy, sensitivity, and specificity respectively to detect MGMT methylation status for patients suffering from glioblastoma. Deep learning feature extraction with radiogenomic features, fusing imaging phenotypes and molecular structure, using rejection algorithm has been found to perform outclass capable of detecting MGMT methylation status of glioblastoma patients. The approach relates the genomic variation with radiomic features forming a bridge between two areas of research that may prove useful for clinical treatment planning leading to better outcomes.
Collapse
Affiliation(s)
- Shahzad Ahmad Qureshi
- Department of Computer and Information Sciences, Pakistan Institute of Engineering and Applied Sciences, Islamabad, Pakistan.
| | - Lal Hussain
- Department of Computer Science and IT, Neelum Campus, The University of Azad Jammu and Kashmir, Muzaffarabad, Azad Kashmir, Pakistan. .,Department of Computer Science and IT, King Abdullah Campus, The University of Azad Jammu and Kashmir, Muzaffarabad, Azad Kashmir, Pakistan. .,Department of Radiology, Albert Einstein College of Medicine and Montefiore Medical Center, 111 East 210th Street, Bronx, NY, 10467, USA.
| | - Usama Ibrar
- grid.461150.7Farooq Hospital, Lahore, Pakistan
| | - Eatedal Alabdulkreem
- grid.449346.80000 0004 0501 7602Department of Computer Sciences, College of Computer and Information Sciences, Princess Nourah Bint Abdulrahman University, P.O. Box 84428, Riyadh, 11671 Saudi Arabia
| | - Mohamed K. Nour
- grid.412832.e0000 0000 9137 6644Department of Computer Sciences, College of Computing and Information System, Umm Al-Qura University, Mecca, Saudi Arabia
| | - Mohammed S. Alqahtani
- grid.412144.60000 0004 1790 7100Radiological Sciences Department, College of Applied Medical Sciences, King Khalid University, Abha, 61421 Saudi Arabia
| | - Faisal Mohammed Nafie
- grid.449051.d0000 0004 0441 5633Department of Computer Science, College of Science and Humanities at Alghat, Majmaah University, Al-Majmaah, 11952 Saudi Arabia
| | - Abdullah Mohamed
- grid.440865.b0000 0004 0377 3762Research Centre, Future University in Egypt, New Cairo, 11845 Egypt
| | - Gouse Pasha Mohammed
- grid.449553.a0000 0004 0441 5588Department of Computer and Self Development, Preparatory Year Deanship, Prince Sattam Bin Abdulaziz University, AlKharj, Saudi Arabia
| | - Tim Q. Duong
- grid.240283.f0000 0001 2152 0791Department of Radiology, Albert Einstein College of Medicine and Montefiore Medical Center, 111 East 210th Street, Bronx, NY 10467 USA
| |
Collapse
|
16
|
Sha Y, Yan Q, Tan Y, Wang X, Zhang H, Yang G. Prediction of the Molecular Subtype of IDH Mutation Combined with MGMT Promoter Methylation in Gliomas via Radiomics Based on Preoperative MRI. Cancers (Basel) 2023; 15:cancers15051440. [PMID: 36900232 PMCID: PMC10001198 DOI: 10.3390/cancers15051440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 02/12/2023] [Accepted: 02/21/2023] [Indexed: 03/12/2023] Open
Abstract
BACKGROUND The molecular subtype of IDH mut combined with MGMT meth in gliomas suggests a good prognosis and potential benefit from TMZ chemotherapy. The aim of this study was to establish a radiomics model to predict this molecular subtype. METHOD The preoperative MR images and genetic data of 498 patients with gliomas were retrospectively collected from our institution and the TCGA/TCIA dataset. A total of 1702 radiomics features were extracted from the tumour region of interest (ROI) of CE-T1 and T2-FLAIR MR images. Least absolute shrinkage and selection operator (LASSO) and logistic regression were used for feature selection and model building. Receiver operating characteristic (ROC) curves and calibration curves were used to evaluate the predictive performance of the model. RESULTS Regarding clinical variables, age and tumour grade were significantly different between the two molecular subtypes in the training, test and independent validation cohorts (p < 0.05). The areas under the curve (AUCs) of the radiomics model based on 16 selected features in the SMOTE training cohort, un-SMOTE training cohort, test set and independent TCGA/TCIA validation cohort were 0.936, 0.932, 0.916 and 0.866, respectively, and the corresponding F1-scores were 0.860, 0.797, 0.880 and 0.802. The AUC of the independent validation cohort increased to 0.930 for the combined model when integrating the clinical risk factors and radiomics signature. CONCLUSIONS radiomics based on preoperative MRI can effectively predict the molecular subtype of IDH mut combined with MGMT meth.
Collapse
Affiliation(s)
- Yongjian Sha
- Department of Radiology, First Hospital of Shanxi Medical University, Taiyuan 030001, China
- Xi'an No.3 Hospital, Affiliated Hospital of Northwest University, Xi'an 710018, China
| | - Qianqian Yan
- Department of Radiology, First Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Yan Tan
- Department of Radiology, First Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Xiaochun Wang
- Department of Radiology, First Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Hui Zhang
- Department of Radiology, First Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Guoqiang Yang
- Department of Radiology, First Hospital of Shanxi Medical University, Taiyuan 030001, China
| |
Collapse
|
17
|
Saxena S, Jena B, Mohapatra B, Gupta N, Kalra M, Scartozzi M, Saba L, Suri JS. Fused deep learning paradigm for the prediction of o6-methylguanine-DNA methyltransferase genotype in glioblastoma patients: A neuro-oncological investigation. Comput Biol Med 2023; 153:106492. [PMID: 36621191 DOI: 10.1016/j.compbiomed.2022.106492] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 11/29/2022] [Accepted: 12/27/2022] [Indexed: 01/06/2023]
Abstract
BACKGROUND The O6-methylguanine-DNA methyltransferase (MGMT) is a deoxyribonucleic acid (DNA) repairing enzyme that has been established as an essential clinical brain tumor biomarker for Glioblastoma Multiforme (GBM). Knowing the status of MGMT methylation biomarkers using multi-parametric MRI (mp-MRI) helps neuro-oncologists to analyze GBM and its treatment plan. METHOD The hand-crafted radiomics feature extraction of GBM's subregions, such as edema(ED), tumor core (TC), and enhancing tumor (ET) in the machine learning (ML) framework, was investigated using support vector machine(SVM), K-Nearest Neighbours (KNN), random forest (RF), LightGBM, and extreme gradient boosting (XGB). For tissue-level analysis of the promotor genes in GBM, we used the deep residual neural network (ResNet-18) with 3D architecture, followed by EfficientNet-based investigation for variants as B0 and B1. Lastly, we analyzed the fused deep learning (FDL) framework that combines ML and DL frameworks. RESULT Structural mp-MRI consisting of T1, T2, FLAIR, and T1GD having a size of 400 and 185 patients, respectively, for discovery and replication cohorts. Using the CV protocol in the ResNet-3D framework, MGMT methylation status prediction in mp-MRI gave the AUC of 0.753 (p < 0.0001) and 0.72 (p < 0.0001) for the discovery and replication cohort, respectively. We presented that the FDL is ∼7% superior to solo DL and ∼15% to solo ML. CONCLUSION The proposed study aims to provide solutions for building an efficient predictive model of MGMT for GBM patients using deep radiomics features obtained from mp-MRI with the end-to-end ResNet-18 3D and FDL imaging signatures.
Collapse
Affiliation(s)
- Sanjay Saxena
- Department of Computer Science & Engineering, International Institute of Information Technology, Bhubaneswar, Odisha, India
| | - Biswajit Jena
- Department of Computer Science & Engineering, Institute of Technical Education and Research, SOA Deemed to be University, Bhubaneswar, India
| | - Bibhabasu Mohapatra
- Department of Computer Science & Engineering, International Institute of Information Technology, Bhubaneswar, Odisha, India
| | - Neha Gupta
- Bharati Vidyapeeth's College of Engineering, Paschim Vihar, New Delhi, India
| | - Manudeep Kalra
- Department of Radiology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Mario Scartozzi
- Department of Radiology, A.O.U, di Cagliari-Polo di Monserrato s.s, 09124, Cagliari, Italy
| | - Luca Saba
- Department of Radiology, A.O.U, di Cagliari-Polo di Monserrato s.s, 09124, Cagliari, Italy
| | - Jasjit S Suri
- Stroke Monitoring and Diagnostic Division, AtheroPoint™ LLC, Roseville, CA, USA; Knowledge Engineering Centre, Global Biomedical Technologies, Inc, Roseville, CA, USA.
| |
Collapse
|
18
|
Liu S, Zhang Y, Kong Z, Jiang C, Wang Y, Zhao D, You H, Ma W, Feng F. Feasibility of evaluating the histologic and genetic subtypes of WHO grade II-IV gliomas by diffusion-weighted imaging. BMC Neurosci 2022; 23:72. [PMID: 36471242 PMCID: PMC9720933 DOI: 10.1186/s12868-022-00750-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 10/28/2022] [Indexed: 12/07/2022] Open
Abstract
BACKGROUND To explore the feasibility of diffusion-weighted imaging (DWI) metrics to predict the histologic subtypes and genetic status of gliomas (e.g., IDH, MGMT, and TERT) noninvasively. METHODS One hundred and eleven patients with pathologically confirmed WHO grade II-IV gliomas were recruited retrospectively. Apparent diffusion coefficient (ADC) values were measured in solid parts of gliomas on co-registered T2-weighted images and were compared with each other in terms of WHO grading and genotypes using t-tests. Receiver operating characteristic analysis was performed to assess the diagnostic performances of ADC. Subsequently, multiple linear regression was used to find independent variables, which can directly affect ADC values. RESULTS The values of overall mean ADC (omADC) and normalized ADC (nADC) of high grade gliomas and IDH wildtype gliomas were lower than low grade gliomas and IDH mutated gliomas (P < 0.05). nADC values showed better diagnostic performance than omADC in identifying tumor grade (AUC: 0.787 vs. 0.750) and IDH status (AUC: 0.836 vs. 0.777). ADC values had limited abilities in distinguishing TERT status (AUC = 0.607 for nADC and 0.617 for omADC) and MGMT status (AUC = 0.651 for nADC). Only tumor grade and IDH status were tightly associated with ADC values. CONCLUSION DWI metrics can predict glioma grading and IDH mutation noninvasively, but have limited use in detecting TERT mutation and MGMT methylation.
Collapse
Affiliation(s)
- Sirui Liu
- grid.506261.60000 0001 0706 7839Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China ,grid.8547.e0000 0001 0125 2443Department of Radiology, Shanghai Institute of Medical Imaging, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yiwei Zhang
- grid.506261.60000 0001 0706 7839Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China ,grid.411472.50000 0004 1764 1621Department of Radiology, Peking University First Hospital, No.8 Xishiku, Beijing, China
| | - Ziren Kong
- grid.506261.60000 0001 0706 7839Department of Neurosurgery, Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China
| | - Chendan Jiang
- grid.506261.60000 0001 0706 7839Department of Neurosurgery, Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China
| | - Yu Wang
- grid.506261.60000 0001 0706 7839Department of Neurosurgery, Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China
| | - Dachun Zhao
- grid.506261.60000 0001 0706 7839Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hui You
- grid.506261.60000 0001 0706 7839Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China
| | - Wenbin Ma
- grid.506261.60000 0001 0706 7839Department of Neurosurgery, Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China
| | - Feng Feng
- grid.506261.60000 0001 0706 7839Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China
| |
Collapse
|
19
|
Improving MGMT methylation status prediction of glioblastoma through optimizing radiomics features using genetic algorithm-based machine learning approach. Sci Rep 2022; 12:13412. [PMID: 35927323 PMCID: PMC9352871 DOI: 10.1038/s41598-022-17707-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 07/29/2022] [Indexed: 11/30/2022] Open
Abstract
O6-Methylguanine-DNA-methyltransferase (MGMT) promoter methylation was shown in many studies to be an important predictive biomarker for temozolomide (TMZ) resistance and poor progression-free survival in glioblastoma multiforme (GBM) patients. However, identifying the MGMT methylation status using molecular techniques remains challenging due to technical limitations, such as the inability to obtain tumor specimens, high prices for detection, and the high complexity of intralesional heterogeneity. To overcome these difficulties, we aimed to test the feasibility of using a novel radiomics-based machine learning (ML) model to preoperatively and noninvasively predict the MGMT methylation status. In this study, radiomics features extracted from multimodal images of GBM patients with annotated MGMT methylation status were downloaded from The Cancer Imaging Archive (TCIA) public database for retrospective analysis. The radiomics features extracted from multimodal images from magnetic resonance imaging (MRI) had undergone a two-stage feature selection method, including an eXtreme Gradient Boosting (XGBoost) feature selection model followed by a genetic algorithm (GA)-based wrapper model for extracting the most meaningful radiomics features for predictive purposes. The cross-validation results suggested that the GA-based wrapper model achieved the high performance with a sensitivity of 0.894, specificity of 0.966, and accuracy of 0.925 for predicting the MGMT methylation status in GBM. Application of the extracted GBM radiomics features on a low-grade glioma (LGG) dataset also achieved a sensitivity 0.780, specificity 0.620, and accuracy 0.750, indicating the potential of the selected radiomics features to be applied more widely on both low- and high-grade gliomas. The performance indicated that our model may potentially confer significant improvements in prognosis and treatment responses in GBM patients.
Collapse
|
20
|
Zheng X, Xu S, Wu J. Cervical Cancer Imaging Features Associated With ADRB1 as a Risk Factor for Cerebral Neurovascular Metastases. Front Neurol 2022; 13:905761. [PMID: 35903112 PMCID: PMC9315067 DOI: 10.3389/fneur.2022.905761] [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: 03/27/2022] [Accepted: 06/20/2022] [Indexed: 11/13/2022] Open
Abstract
Bioinformatics tools are used to create a clinical prediction model for cervical cancer metastasis and to investigate the neurovascular-related genes that are involved in brain metastasis of cervical cancer. One hundred eighteen patients with cervical cancer were divided into two groups based on the presence or absence of metastases, and the clinical data and imaging findings of the two groups were compared retrospectively. The nomogram-based model was successfully constructed by taking into account four clinical characteristics (age, stage, N, and T) as well as one imaging characteristic (original_glszm_GrayLevelVariance Rad-score). In patients with cervical cancer, headaches and vomiting were more often reported in the brain metastasis group than in the other metastasis groups. According to the TCGA data, mRNA differential gene expression analysis of patients with cervical cancer revealed an increase in the expression of neurovascular-related gene Adrenoceptor Beta 1 (ADRB1) in the brain metastasis group. An analysis of the correlation between imaging features and ADRB1 expression revealed that ADRB1 expression was significantly higher in the low Rad-score group compared with the high Rad-score group (P = 0.025). Therefore, ADRB1 expression in cervical cancer was correlated with imaging features and was associated as a risk factor for cerebral neurovascular metastases. This study developed a nomogram prediction model for cervical cancer metastasis using age, stage, N, T and original_glszm_GrayLevelVariance. As a risk factor associated with the development of cerebral neurovascular metastases of cervical cancer, ADRB1 expression was significantly higher in brain metastases from cervical cancer.
Collapse
Affiliation(s)
- Xingju Zheng
- Department of Radiology, Guizhou Provincial People's Hospital, Guiyang, China
| | - Shilin Xu
- Department of Oncology, Xichang People's Hospital, Liangshan High-Tech Tumor Hospital, Xichang, China
| | - JiaYing Wu
- Department of Gynaecology and Obstetrics, Zhejiang Xinda Hospital, Huzhou, China
- *Correspondence: JiaYing Wu
| |
Collapse
|
21
|
A Survey of Radiomics in Precision Diagnosis and Treatment of Adult Gliomas. J Clin Med 2022; 11:jcm11133802. [PMID: 35807084 PMCID: PMC9267404 DOI: 10.3390/jcm11133802] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 06/18/2022] [Accepted: 06/29/2022] [Indexed: 02/04/2023] Open
Abstract
Glioma is the most common primary malignant tumor of the adult central nervous system (CNS), which mostly shows invasive growth. In most cases, surgery is often difficult to completely remove, and the recurrence rate and mortality of patients are high. With the continuous development of molecular genetics and the great progress of molecular biology technology, more and more molecular biomarkers have been proved to have important guiding significance in the individualized diagnosis, treatment, and prognosis evaluation of glioma. With the updates of the World Health Organization (WHO) classification of tumors of the CNS in 2021, the diagnosis and treatment of glioma has entered the era of precision medicine in the true sense. Due to its ability to non-invasively achieve accurate identification of glioma from other intracranial tumors, and to predict the grade, genotyping, treatment response, and prognosis of glioma, which provides a scientific basis for the clinical application of individualized diagnosis and treatment model of glioma, radiomics has become a research hotspot in the field of precision medicine. This paper reviewed the research related to radiomics of adult gliomas published in recent years and summarized the research proceedings of radiomics in differential diagnosis, preoperative grading and genotyping, treatment and efficacy evaluation, and survival prediction of adult gliomas.
Collapse
|
22
|
Chen S, Xu Y, Ye M, Li Y, Sun Y, Liang J, Lu J, Wang Z, Zhu Z, Zhang X, Zhang B. Predicting MGMT Promoter Methylation in Diffuse Gliomas Using Deep Learning with Radiomics. J Clin Med 2022; 11:jcm11123445. [PMID: 35743511 PMCID: PMC9224690 DOI: 10.3390/jcm11123445] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 06/08/2022] [Accepted: 06/13/2022] [Indexed: 02/01/2023] Open
Abstract
This study aimed to investigate the feasibility of predicting oxygen 6-methylguanine-DNA methyltransferase (MGMT) promoter methylation in diffuse gliomas by developing a deep learning approach using MRI radiomics. A total of 111 patients with diffuse gliomas participated in the retrospective study (56 patients with MGMT promoter methylation and 55 patients with MGMT promoter unmethylation). The radiomics features of the two regions of interest (ROI) (the whole tumor area and the tumor core area) for four sequences, including T1 weighted image (T1WI), T2 weighted image (T2WI), apparent diffusion coefficient (ADC) maps, and T1 contrast-enhanced (T1CE) MR images were extracted and jointly fed into the residual network. Then the deep learning method was developed and evaluated with a five-fold cross-validation, where in each fold, the dataset was randomly divided into training (80%) and validation (20%) cohorts. We compared the performance of all models using area under the curve (AUC) and average accuracy of validation cohorts and calculated the 10 most important features of the best model via a class activation map. Based on the ROI of the whole tumor, the predictive capacity of the T1CE and ADC model achieved the highest AUC value of 0.85. Based on the ROI of the tumor core, the T1CE and ADC model achieved the highest AUC value of 0.90. After comparison, the T1CE combined with the ADC model based on the ROI of the tumor core exhibited the best performance, with the highest average accuracy (0.91) and AUC (0.90) among all models. The deep learning method using MRI radiomics has excellent diagnostic performance with a high accuracy in predicting MGMT promoter methylation in diffuse gliomas.
Collapse
Affiliation(s)
- Sixuan Chen
- Department of Radiology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China; (S.C.); (M.Y.); (Y.L.); (J.L.); (Z.W.); (Z.Z.); (B.Z.)
| | - Yue Xu
- National Institute of Healthcare Data Science, Nanjing University, Nanjing 210023, China;
| | - Meiping Ye
- Department of Radiology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China; (S.C.); (M.Y.); (Y.L.); (J.L.); (Z.W.); (Z.Z.); (B.Z.)
| | - Yang Li
- Department of Radiology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China; (S.C.); (M.Y.); (Y.L.); (J.L.); (Z.W.); (Z.Z.); (B.Z.)
| | - Yu Sun
- School of Biological Science and Medical Engineering, Southeast University, Nanjing 211189, China; (Y.S.); (J.L.)
| | - Jiawei Liang
- School of Biological Science and Medical Engineering, Southeast University, Nanjing 211189, China; (Y.S.); (J.L.)
| | - Jiaming Lu
- Department of Radiology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China; (S.C.); (M.Y.); (Y.L.); (J.L.); (Z.W.); (Z.Z.); (B.Z.)
| | - Zhengge Wang
- Department of Radiology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China; (S.C.); (M.Y.); (Y.L.); (J.L.); (Z.W.); (Z.Z.); (B.Z.)
| | - Zhengyang Zhu
- Department of Radiology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China; (S.C.); (M.Y.); (Y.L.); (J.L.); (Z.W.); (Z.Z.); (B.Z.)
| | - Xin Zhang
- Department of Radiology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China; (S.C.); (M.Y.); (Y.L.); (J.L.); (Z.W.); (Z.Z.); (B.Z.)
- Correspondence:
| | - Bing Zhang
- Department of Radiology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China; (S.C.); (M.Y.); (Y.L.); (J.L.); (Z.W.); (Z.Z.); (B.Z.)
- Institute of Brain Science, Nanjing University, Nanjing 210023, China
| |
Collapse
|
23
|
Wu W, Wang Y, Xiang J, Li X, Wahafu A, Yu X, Bai X, Yan G, Wang C, Wang N, Du C, Xie W, Wang M, Wang J. A Novel Multi-Omics Analysis Model for Diagnosis and Survival Prediction of Lower-Grade Glioma Patients. Front Oncol 2022; 12:729002. [PMID: 35646656 PMCID: PMC9133344 DOI: 10.3389/fonc.2022.729002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 03/24/2022] [Indexed: 01/13/2023] Open
Abstract
Background Lower-grade gliomas (LGGs) are characterized by remarkable genetic heterogeneity and different clinical outcomes. Classification of LGGs is improved by the development of molecular stratification markers including IDH mutation and 1p/19q chromosomal integrity, which are used as a hallmark of survival and therapy sensitivity of LGG patients. However, the reproducibility and sensitivity of the current classification remain ambiguous. This study aimed to construct more accurate risk-stratification approaches. Methods According to bioinformatics, the sequencing profiles of methylation and transcription and imaging data derived from LGG patients were analyzed and developed predictable risk score and radiomics score. Moreover, the performance of predictable models was further validated. Results In this study, we determined a cluster of 6 genes that were correlated with IDH mutation/1p19q co-deletion status. Risk score model was calculated based on 6 genes and showed gratifying sensitivity and specificity for survival prediction and therapy response of LGG patients. Furthermore, a radiomics risk score model was established to noninvasively assist judgment of risk score in pre-surgery. Taken together, a predictable nomogram that combined transcriptional signatures and clinical characteristics was established and validated to be preferable to the histopathological classification. Our novel multi-omics nomograms showed a satisfying performance. To establish a user-friendly application, the nomogram was further developed into a web-based platform: https://drw576223193.shinyapps.io/Nomo/, which could be used as a supporting method in addition to the current histopathological-based classification of gliomas. Conclusions Our novel multi-omics nomograms showed the satisfying performance of LGG patients and assisted clinicians to draw up individualized clinical management.
Collapse
Affiliation(s)
- Wei Wu
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Center of Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yichang Wang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Center of Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jianyang Xiang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Center of Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaodong Li
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Center of Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Alafate Wahafu
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Center of Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiao Yu
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaobin Bai
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ge Yan
- Department of Medical Imaging, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Chunbao Wang
- Department of Pathology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ning Wang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Changwang Du
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Wanfu Xie
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Maode Wang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Center of Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jia Wang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Center of Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
24
|
Balana C, Castañer S, Carrato C, Moran T, Lopez-Paradís A, Domenech M, Hernandez A, Puig J. Preoperative Diagnosis and Molecular Characterization of Gliomas With Liquid Biopsy and Radiogenomics. Front Neurol 2022; 13:865171. [PMID: 35693015 PMCID: PMC9177999 DOI: 10.3389/fneur.2022.865171] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 05/05/2022] [Indexed: 12/13/2022] Open
Abstract
Gliomas are a heterogenous group of central nervous system tumors with different outcomes and different therapeutic needs. Glioblastoma, the most common subtype in adults, has a very poor prognosis and disabling consequences. The World Health Organization (WHO) classification specifies that the typing and grading of gliomas should include molecular markers. The molecular characterization of gliomas has implications for prognosis, treatment planning, and prediction of treatment response. At present, gliomas are diagnosed via tumor resection or biopsy, which are always invasive and frequently risky methods. In recent years, however, substantial advances have been made in developing different methods for the molecular characterization of tumors through the analysis of products shed in body fluids. Known as liquid biopsies, these analyses can potentially provide diagnostic and prognostic information, guidance on choice of treatment, and real-time information on tumor status. In addition, magnetic resonance imaging (MRI) is another good source of tumor data; radiomics and radiogenomics can link the imaging phenotypes to gene expression patterns and provide insights to tumor biology and underlying molecular signatures. Machine and deep learning and computational techniques can also use quantitative imaging features to non-invasively detect genetic mutations. The key molecular information obtained with liquid biopsies and radiogenomics can be useful not only in the diagnosis of gliomas but can also help predict response to specific treatments and provide guidelines for personalized medicine. In this article, we review the available data on the molecular characterization of gliomas using the non-invasive methods of liquid biopsy and MRI and suggest that these tools could be used in the future for the preoperative diagnosis of gliomas.
Collapse
Affiliation(s)
- Carmen Balana
- Medical Oncology Service, Institut Català d'Oncologia Badalona (ICO), Badalona Applied Research Group in Oncology (B-ARGO Group), Institut Investigació Germans Trias i Pujol (IGTP), Barcelona, Spain
- *Correspondence: Carmen Balana
| | - Sara Castañer
- Diagnostic Imaging Institute (IDI), Hospital Universitari Germans Trias I Pujol, Institut Investigació Germans Trias i Pujol (IGTP), Barcelona, Spain
| | - Cristina Carrato
- Department of Pathology, Hospital Universitari Germans Trias I Pujol, Institut Investigació Germans Trias i Pujol (IGTP), Barcelona, Spain
| | - Teresa Moran
- Medical Oncology Service, Institut Català d'Oncologia Badalona (ICO), Badalona Applied Research Group in Oncology (B-ARGO Group), Institut Investigació Germans Trias i Pujol (IGTP), Barcelona, Spain
| | - Assumpció Lopez-Paradís
- Medical Oncology Service, Institut Català d'Oncologia Badalona (ICO), Badalona Applied Research Group in Oncology (B-ARGO Group), Institut Investigació Germans Trias i Pujol (IGTP), Barcelona, Spain
| | - Marta Domenech
- Medical Oncology Service, Institut Català d'Oncologia Badalona (ICO), Badalona Applied Research Group in Oncology (B-ARGO Group), Institut Investigació Germans Trias i Pujol (IGTP), Barcelona, Spain
| | - Ainhoa Hernandez
- Medical Oncology Service, Institut Català d'Oncologia Badalona (ICO), Badalona Applied Research Group in Oncology (B-ARGO Group), Institut Investigació Germans Trias i Pujol (IGTP), Barcelona, Spain
| | - Josep Puig
- Department of Radiology IDI [Girona Biomedical Research Institute] IDIBGI, Hospital Universitari Dr Josep Trueta, Girona, Spain
- Department of Medical Sciences, School of Medicine, University of Girona, Girona, Spain
- Comparative Medicine and Bioimage of Catalonia, Institut Investigació Germans Trias i Pujol (IGTP), Barcelona, Spain
| |
Collapse
|
25
|
A radiomics feature-based nomogram to predict telomerase reverse transcriptase promoter mutation status and the prognosis of lower-grade gliomas. Clin Radiol 2022; 77:e560-e567. [PMID: 35595562 DOI: 10.1016/j.crad.2022.04.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 04/07/2022] [Indexed: 11/21/2022]
Abstract
AIM To explore the predictive value of the radiomics feature-based nomogram for predicting telomerase reverse transcriptase (TERT) promoter mutation status and prognosis of lower-grade gliomas (LGGs) non-invasively. MATERIALS AND METHODS One hundred and seventy-six LGG patients (123 in the training cohort and 53 in the validation cohort) were enrolled retrospectively. A total of 851 radiomics features were extracted from contrast-enhanced magnetic resonance imaging (MRI) images. The radiomics features were selected using the least absolute shrinkage and selection operator (LASSO) method and a rad-score was calculated. Multivariate logistic regression analysis was used to build a radiomics signature based on rad-score, participant's age, and gender, and a radiomics nomogram was used to represent this signature. The performance of the signature was evaluated by receiver operating characteristic (ROC) curve analysis, and the patient prognosis was stratified based on the TERT promoter mutation status and the radiomics signature. RESULTS Seven robust radiomics features were selected by LASSO and the radiomics signature showed good performance for predicting the TERT promoter mutation status, with an area under the curve (AUC) of 0.900 (0.832-0.946) and 0.873 (0.753-0.948) in the training and validation datasets. With a median overall survival time of 28.5 months, the radiomics signature stratified the LGG patients into two risk groups with significantly different prognosis (log-rank = 47.531, p<0.001). CONCLUSION The radiomics feature-based nomogram is a promising approach for predicting the TERT promoter mutation status preoperatively and evaluating the prognosis of lower-grade glioma patients non-invasively.
Collapse
|
26
|
Deng DB, Liao YT, Zhou JF, Cheng LN, He P, Wu SN, Wang WS, Zhou Q. Non-Invasive Prediction of Survival Time of Midline Glioma Patients Using Machine Learning on Multiparametric MRI Radiomics Features. Front Neurol 2022; 13:866274. [PMID: 35585843 PMCID: PMC9108285 DOI: 10.3389/fneur.2022.866274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/31/2022] [Indexed: 11/22/2022] Open
Abstract
OBJECTIVES To explore the feasibility of predicting overall survival (OS) of patients with midline glioma using multi-parameter magnetic resonance imaging (MRI) features. METHODS Data of 84 patients with midline gliomas were retrospectively collected, including 40 patients with OS > 12 months (28 cases were adults, 14 cases were H3 K27M-mutation) and 44 patients with OS < 12 months (29 cases were adults, 31 cases were H3 K27M-mutation). Features were extracted from the largest slice of tumors, which were manually segmented on T2-weighted (T2w), T2 fluid-attenuated inversion recovery (T2 FLAIR), and contrast-enhanced T1-weighted (T1c) images. Data were randomly divided into training (70%) and test cohorts (30%) and normalized and standardized using Z-scores. Feature dimensionality reduction was performed using the variance method and maximum relevance and minimum redundancy (mRMR) algorithm. We used the logistic regression algorithm to construct three models for T2w, T2 FLAIR, and T1c images as well as one combined model. The test cohort was used to evaluate the models, and receiver operating characteristic (ROC) curves, areas under the curve (AUCs), sensitivity, specificity, and accuracy were calculated. The nomogram of the combined model was built and evaluated using a calibration curve. Decision curve analysis (DCA) was used to evaluate the clinical application value of the four models. RESULTS A total of 1,316 features were extracted from T2w, T2 FLAIR, and T1c images, respectively. And then the best non-redundant features were selected from the extracted features using the variance method and mRMR. Finally, five features were extracted each from T2w, T2 FLAIR, and T1c images, and 12 features were extracted for the combined model. Four models were established using the optimal features. In the test cohort, the combined model performed the best out of all models. The AUCs of the T2w, T2 FLAIR, T1c, and combined models were 0.73, 0.78, 0.74, and 0.87, respectively, and accuracies were 0.72, 0.76, 0.72, and 0.84, respectively. The ROC curves and DCA showed that the combined model had the highest efficiency and most favorable clinical benefits. CONCLUSION The combined radiomics model based on multi-parameter MRI features provided a reliable non-invasive method for the prognostic prediction of midline gliomas.
Collapse
Affiliation(s)
- Da-Biao Deng
- Department of Radiology, Third Affiliated Hospital of Southern Medical University (Academy of Orthopedics Guangdong), Guangzhou, China
- Imaging Department of Guangdong 999 Brain Hospital, Guangzhou, China
| | | | - Jiang-Fen Zhou
- Department of Neuro-Oncology of Guangdong 999 Brain Hospital, Guangzhou, China
| | - Li-Na Cheng
- Imaging Department of Guangdong 999 Brain Hospital, Guangzhou, China
| | - Peng He
- Imaging Department of Guangdong 999 Brain Hospital, Guangzhou, China
| | - Sheng-Nan Wu
- Imaging Department of Guangdong 999 Brain Hospital, Guangzhou, China
| | - Wen-Sheng Wang
- Imaging Department of Guangdong 999 Brain Hospital, Guangzhou, China
| | - Quan Zhou
- Department of Radiology, Third Affiliated Hospital of Southern Medical University (Academy of Orthopedics Guangdong), Guangzhou, China
| |
Collapse
|
27
|
Overall Survival Prognostic Modelling of Non-small Cell Lung Cancer Patients Using Positron Emission Tomography/Computed Tomography Harmonised Radiomics Features: The Quest for the Optimal Machine Learning Algorithm. Clin Oncol (R Coll Radiol) 2021; 34:114-127. [PMID: 34872823 DOI: 10.1016/j.clon.2021.11.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/01/2021] [Accepted: 11/17/2021] [Indexed: 02/06/2023]
Abstract
AIMS Despite the promising results achieved by radiomics prognostic models for various clinical applications, multiple challenges still need to be addressed. The two main limitations of radiomics prognostic models include information limitation owing to single imaging modalities and the selection of optimum machine learning and feature selection methods for the considered modality and clinical outcome. In this work, we applied several feature selection and machine learning methods to single-modality positron emission tomography (PET) and computed tomography (CT) and multimodality PET/CT fusion to identify the best combinations for different radiomics modalities towards overall survival prediction in non-small cell lung cancer patients. MATERIALS AND METHODS A PET/CT dataset from The Cancer Imaging Archive, including subjects from two independent institutions (87 and 95 patients), was used in this study. Each cohort was used once as training and once as a test, followed by averaging of the results. ComBat harmonisation was used to address the centre effect. In our proposed radiomics framework, apart from single-modality PET and CT models, multimodality radiomics models were developed using multilevel (feature and image levels) fusion. Two different methods were considered for the feature-level strategy, including concatenating PET and CT features into a single feature set and alternatively averaging them. For image-level fusion, we used three different fusion methods, namely wavelet fusion, guided filtering-based fusion and latent low-rank representation fusion. In the proposed prognostic modelling framework, combinations of four feature selection and seven machine learning methods were applied to all radiomics modalities (two single and five multimodalities), machine learning hyper-parameters were optimised and finally the models were evaluated in the test cohort with 1000 repetitions via bootstrapping. Feature selection and machine learning methods were selected as popular techniques in the literature, supported by open source software in the public domain and their ability to cope with continuous time-to-event survival data. Multifactor ANOVA was used to carry out variability analysis and the proportion of total variance explained by radiomics modality, feature selection and machine learning methods was calculated by a bias-corrected effect size estimate known as ω2. RESULTS Optimum feature selection and machine learning methods differed owing to the applied radiomics modality. However, minimum depth (MD) as feature selection and Lasso and Elastic-Net regularized generalized linear model (glmnet) as machine learning method had the highest average results. Results from the ANOVA test indicated that the variability that each factor (radiomics modality, feature selection and machine learning methods) introduces to the performance of models is case specific, i.e. variances differ regarding different radiomics modalities and fusion strategies. Overall, the greatest proportion of variance was explained by machine learning, except for models in feature-level fusion strategy. CONCLUSION The identification of optimal feature selection and machine learning methods is a crucial step in developing sound and accurate radiomics risk models. Furthermore, optimum methods are case specific, differing due to the radiomics modality and fusion strategy used.
Collapse
|
28
|
Huang H, Wang FF, Luo S, Chen G, Tang G. Diagnostic performance of radiomics using machine learning algorithms to predict MGMT promoter methylation status in glioma patients: a meta-analysis. DIAGNOSTIC AND INTERVENTIONAL RADIOLOGY (ANKARA, TURKEY) 2021; 27:716-724. [PMID: 34792025 DOI: 10.5152/dir.2021.21153] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE We aimed to assess the diagnostic performance of radiomics using machine learning algorithms to predict the methylation status of the O6-methylguanine-DNA methyltransferase (MGMT) promoter in glioma patients. METHODS A comprehensive literature search of PubMed, EMBASE, and Web of Science until 27 July 2021 was performed to identify eligible studies. Stata SE 15.0 and Meta-Disc 1.4 were used for data analysis. RESULTS A total of fifteen studies with 1663 patients were included: five studies with training and validation cohorts and ten with only training cohorts. The pooled sensitivity and specificity of machine learning for predicting MGMT promoter methylation in gliomas were 85% (95% CI 79%-90%) and 84% (95% CI 78%-88%) in the training cohort (n=15) and 84% (95% CI 70%-92%) and 78% (95% CI 63%-88%) in the validation cohort (n=5). The AUC was 0.91 (95% CI 0.88-0.93) in the training cohort and 0.88 (95% CI 0.85-0.91) in the validation cohort. The meta-regression demonstrated that magnetic resonance imaging sequences were related to heterogeneity. The sensitivity analysis showed that heterogeneity was reduced by excluding one study with the lowest diagnostic performance. CONCLUSION This meta-analysis demonstrated that machine learning is a promising, reliable and repeatable candidate method for predicting MGMT promoter methylation status in glioma and showed a higher performance than non-machine learning methods.
Collapse
Affiliation(s)
- Huan Huang
- Department of Radiology, Affiliated Hospital of Southwest Medical University, Sichuan, China
| | - Fei-Fei Wang
- Department of Radiology, Affiliated Hospital of Southwest Medical University, Sichuan, China
| | - Shigang Luo
- Department of Radiology, Affiliated Hospital of Southwest Medical University, Sichuan, China
| | - Guangxiang Chen
- Department of Radiology, Affiliated Hospital of Southwest Medical University, Sichuan, China
| | - Guangcai Tang
- Department of Radiology, Affiliated Hospital of Southwest Medical University, Sichuan, China
| |
Collapse
|
29
|
Amini M, Nazari M, Shiri I, Hajianfar G, Deevband MR, Abdollahi H, Arabi H, Rahmim A, Zaidi H. Multi-level multi-modality (PET and CT) fusion radiomics: prognostic modeling for non-small cell lung carcinoma. Phys Med Biol 2021; 66. [PMID: 34544053 DOI: 10.1088/1361-6560/ac287d] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 09/20/2021] [Indexed: 12/23/2022]
Abstract
We developed multi-modality radiomic models by integrating information extracted from18F-FDG PET and CT images using feature- and image-level fusions, toward improved prognosis for non-small cell lung carcinoma (NSCLC) patients. Two independent cohorts of NSCLC patients from two institutions (87 and 95 patients) were cycled as training and testing datasets. Fusion approaches were applied at two levels, namely feature- and image-levels. For feature-level fusion, radiomic features were extracted individually from CT and PET images and concatenated. Alternatively, radiomic features extracted separately from CT and PET images were averaged. For image-level fusion, wavelet fusion was utilized and tuned with two parameters, namely CT weight and Wavelet Band Pass Filtering Ratio. Clinical and combined clinical + radiomic models were developed. Gray level discretization was performed at 3 different levels (16, 32 and 64) and 225 radiomics features were extracted. Overall survival (OS) was considered as the endpoint. For feature reduction, correlated (redundant) features were excluded using Spearman's correlation, and best combination of top ten features with highest concordance-indices (via univariate Cox model) were selected in each model for further multivariate Cox model. Moreover, prognostic score's median, obtained from the training cohort, was used intact in the testing cohort as a threshold to classify patients into low- versus high-risk groups, and log-rank test was applied to assess differences between the Kaplan-Meier curves. Overall, while models based on feature-level fusion strategy showed limited superiority over single-modalities, image-level fusion strategy significantly outperformed both single-modality and feature-level fusion strategies. As such, the clinical model (C-index = 0.656) outperformed all models from single-modality and feature-level strategies, but was outperformed by certain models from image-level fusion strategy. Our findings indicated that image-level fusion multi-modality radiomics models outperformed single-modality, feature-level fusion, and clinical models for OS prediction of NSCLC patients.
Collapse
Affiliation(s)
- Mehdi Amini
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, CH-1205 Geneva, Switzerland.,Department of Biomedical Engineering and Medical Physics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mostafa Nazari
- Department of Biomedical Engineering and Medical Physics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Isaac Shiri
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, CH-1205 Geneva, Switzerland
| | - Ghasem Hajianfar
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Science, Tehran, Iran
| | - Mohammad Reza Deevband
- Department of Biomedical Engineering and Medical Physics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamid Abdollahi
- Department of Radiologic Technology, School of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Hossein Arabi
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, CH-1205 Geneva, Switzerland
| | - Arman Rahmim
- Departments of Radiology and Physics, University of British Columbia, Vancouver BC, Canada.,Department of Integrative Oncology, BC Cancer Research Institute, Vancouver BC, Canada
| | - Habib Zaidi
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, CH-1205 Geneva, Switzerland.,Geneva University Neurocenter, Geneva University, CH-1211 Geneva, Switzerland.,Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands.,Department of Nuclear Medicine, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
30
|
Radiomics-based MRI for predicting Erythropoietin-producing hepatocellular receptor A2 expression and tumor grade in brain diffuse gliomas. Neuroradiology 2021; 64:323-331. [PMID: 34368897 DOI: 10.1007/s00234-021-02780-1] [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: 04/13/2021] [Accepted: 07/30/2021] [Indexed: 10/20/2022]
Abstract
PURPOSE EphA2 is a key factor underlying invasive propensity of gliomas, and is associated with poor prognosis of tumors. We aimed to develop a radiomics-based imaging index for predicting EphA2 expression in diffuse gliomas, and further estimating its value for grading of tumors. METHODS A total of 182 patients with diffuse gliomas were included. All subjects underwent pre-operative MRI and post-operative pathological diagnosis. EphA2 expression of tumors was scored on pathological sections with immunohistochemical staining using monoclonal EphA2 antibody. MRI radiomics features were extracted from three-dimensional contrast-enhanced T1-weighted imaging and diffusion kurtosis imaging. Predictive models were constructed using machine learning-based radiomics features selection and three classifiers for predicting EphA2 expression and tumor grade. Features of best EphA2 expression model were subsequently used to construct another model of tumor grading. For each model, 146 cases (80%) were randomly picked as training and the rest 36 (20%) were testing cohorts. EphA2 expression was further correlated to the radiomics features in both grade models using Spearman's correlation. RESULTS Logistic regression model presented highest performance for predicting EphA2 expression (AUC: 0.836/0.724 in training/validation set). Tumor gradings model guided by features from EphA2 expression model demonstrated comparable performance (AUC: 0.930/0.983) to that constructed directly using imaging radiomics features (AUC: 0.960/0.977). Two radiomics features which included in both LR-grade models showed strong correlation (P < 0.05) with EphA2 expression. CONCLUSION The expression of EphA2 in gliomas could be predicted by radiomics features extracted from diffusion kurtosis MRI, which could also be used to assist tumor grading.
Collapse
|
31
|
Kinoshita M, Kanemura Y, Narita Y, Kishima H. Reverse Engineering Glioma Radiomics to Conventional Neuroimaging. Neurol Med Chir (Tokyo) 2021; 61:505-514. [PMID: 34373429 PMCID: PMC8443974 DOI: 10.2176/nmc.ra.2021-0133] [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] [Indexed: 11/20/2022] Open
Abstract
A novel radiological research field pursuing comprehensive quantitative image, namely “Radiomics,” gained traction along with the advancement of computational technology and artificial intelligence. This novel concept for analyzing medical images brought extensive interest to the neuro-oncology and neuroradiology research community to build a diagnostic workflow to detect clinically relevant genetic alteration of gliomas noninvasively. Although quite a few promising results were published regarding MRI-based diagnosis of isocitrate dehydrogenase (IDH) mutation in gliomas, it has become clear that an ample amount of effort is still needed to render this technology clinically applicable. At the same time, many significant insights were discovered through this research project, some of which could be “reverse engineered” to improve conventional non-radiomic MR image acquisition. In this review article, the authors aim to discuss the recent advancements and encountering issues of radiomics, how we can apply the knowledge provided by radiomics to standard clinical images, and further expected technological advances in the realm of radiomics and glioma.
Collapse
Affiliation(s)
- Manabu Kinoshita
- Department of Neurosurgery, Asahikawa Medical University.,Department of Neurosurgery, Osaka University Graduate School of Medicine.,Department of Neurosurgery, Osaka International Cancer Institute
| | - Yonehiro Kanemura
- Department of Biomedical Research and Innovation, Institute for Clinical Research, National Hospital Organization Osaka National Hospital
| | - Yoshitaka Narita
- Department of Neurosurgery and Neuro-Oncology, National Cancer Center Hospital
| | - Haruhiko Kishima
- Department of Neurosurgery, Osaka University Graduate School of Medicine
| |
Collapse
|
32
|
Jafari SH, Rabiei N, Taghizadieh M, Mirazimi SMA, Kowsari H, Farzin MA, Razaghi Bahabadi Z, Rezaei S, Mohammadi AH, Alirezaei Z, Dashti F, Nejati M. Joint application of biochemical markers and imaging techniques in the accurate and early detection of glioblastoma. Pathol Res Pract 2021; 224:153528. [PMID: 34171601 DOI: 10.1016/j.prp.2021.153528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 06/09/2021] [Accepted: 06/14/2021] [Indexed: 11/28/2022]
Abstract
Glioblastoma is a primary brain tumor with the most metastatic effect in adults. Despite the wide range of multidimensional treatments, tumor heterogeneity is one of the main causes of tumor spread and gives great complexity to diagnostic and therapeutic methods. Therefore, featuring noble noninvasive prognostic methods that are focused on glioblastoma heterogeneity is perceived as an urgent need. Imaging neuro-oncological biomarkers including MGMT (O6-methylguanine-DNA methyltransferase) promoter methylation status, tumor grade along with other tumor characteristics and demographic features (e.g., age) are commonly referred to during diagnostic, therapeutic and prognostic processes. Therefore, the use of new noninvasive prognostic methods focused on glioblastoma heterogeneity is considered an urgent need. Some neuronal biomarkers, including the promoter methylation status of the promoter MGMT, the characteristics and grade of the tumor, along with the patient's demographics (such as age and sex) are involved in diagnosis, treatment, and prognosis. Among the wide array of imaging techniques, magnetic resonance imaging combined with the more physiologically detailed technique of H-magnetic resonance spectroscopy can be useful in diagnosing neurological cancer patients. In addition, intracranial tumor qualitative analysis and sometimes tumor biopsies help in accurate diagnosis. This review summarizes the evidence for biochemical biomarkers being a reliable biomarker in the early detection and disease management in GBM. Moreover, we highlight the correlation between Imaging techniques and biochemical biomarkers and ask whether they can be combined.
Collapse
Affiliation(s)
- Seyed Hamed Jafari
- Medical Imaging Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nikta Rabiei
- School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Taghizadieh
- Department of Pathology, School of Medicine, Center for Women's Health Research Zahra, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sayad Mohammad Ali Mirazimi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran; Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Hamed Kowsari
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran; Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Amin Farzin
- Department of Laboratory Medicine, School of Allied Medical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Zahra Razaghi Bahabadi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran; Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Samaneh Rezaei
- Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Hossein Mohammadi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Zahra Alirezaei
- Department of Medical Physics, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran; Paramedical School, Bushehr University of Medical Sciences, Bushehr, Iran.
| | - Fatemeh Dashti
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran; Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran.
| | - Majid Nejati
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
33
|
van Kempen EJ, Post M, Mannil M, Kusters B, ter Laan M, Meijer FJA, Henssen DJHA. Accuracy of Machine Learning Algorithms for the Classification of Molecular Features of Gliomas on MRI: A Systematic Literature Review and Meta-Analysis. Cancers (Basel) 2021; 13:cancers13112606. [PMID: 34073309 PMCID: PMC8198025 DOI: 10.3390/cancers13112606] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/17/2021] [Accepted: 05/18/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Glioma prognosis and treatment are based on histopathological characteristics and molecular profile. Following the World Health Organization (WHO) guidelines (2016), the most important molecular diagnostic markers include IDH1/2-genotype and 1p/19q codeletion status, although more recent publications also include ARTX genotype and TERT- and MGMT promoter methylation. Machine learning algorithms (MLAs), however, were described to successfully determine these molecular characteristics non-invasively by using magnetic resonance imaging (MRI) data. The aim of this review and meta-analysis was to define the diagnostic accuracy of MLAs with regard to these different molecular markers. We found high accuracies of MLAs to predict each individual molecular marker, with IDH1/2-genotype being the most investigated and the most accurate. Radiogenomics could therefore be a promising tool for discriminating genetically determined gliomas in a non-invasive fashion. Although encouraging results are presented here, large-scale, prospective trials with external validation groups are warranted. Abstract Treatment planning and prognosis in glioma treatment are based on the classification into low- and high-grade oligodendroglioma or astrocytoma, which is mainly based on molecular characteristics (IDH1/2- and 1p/19q codeletion status). It would be of great value if this classification could be made reliably before surgery, without biopsy. Machine learning algorithms (MLAs) could play a role in achieving this by enabling glioma characterization on magnetic resonance imaging (MRI) data without invasive tissue sampling. The aim of this study is to provide a performance evaluation and meta-analysis of various MLAs for glioma characterization. Systematic literature search and meta-analysis were performed on the aggregated data, after which subgroup analyses for several target conditions were conducted. This study is registered with PROSPERO, CRD42020191033. We identified 724 studies; 60 and 17 studies were eligible to be included in the systematic review and meta-analysis, respectively. Meta-analysis showed excellent accuracy for all subgroups, with the classification of 1p/19q codeletion status scoring significantly poorer than other subgroups (AUC: 0.748, p = 0.132). There was considerable heterogeneity among some of the included studies. Although promising results were found with regard to the ability of MLA-tools to be used for the non-invasive classification of gliomas, large-scale, prospective trials with external validation are warranted in the future.
Collapse
Affiliation(s)
- Evi J. van Kempen
- Department of Medical Imaging, Radboud University Medical Center, Radboud University, 6500HB Nijmegen, The Netherlands; (E.J.v.K.); (M.P.); (F.J.A.M.)
| | - Max Post
- Department of Medical Imaging, Radboud University Medical Center, Radboud University, 6500HB Nijmegen, The Netherlands; (E.J.v.K.); (M.P.); (F.J.A.M.)
| | - Manoj Mannil
- Clinic of Radiology, University Hospital Münster, WWU University of Münster, 48149 Münster, Germany;
| | - Benno Kusters
- Department of Pathology, Radboud University Medical Center, Radboud University, 6500HB Nijmegen, The Netherlands;
| | - Mark ter Laan
- Department of Neurosurgery, Radboud University Medical Center, Radboud University, 6500HB Nijmegen, The Netherlands;
| | - Frederick J. A. Meijer
- Department of Medical Imaging, Radboud University Medical Center, Radboud University, 6500HB Nijmegen, The Netherlands; (E.J.v.K.); (M.P.); (F.J.A.M.)
| | - Dylan J. H. A. Henssen
- Department of Medical Imaging, Radboud University Medical Center, Radboud University, 6500HB Nijmegen, The Netherlands; (E.J.v.K.); (M.P.); (F.J.A.M.)
- Correspondence:
| |
Collapse
|
34
|
Huang WY, Wen LH, Wu G, Pang PP, Ogbuji R, Zhang CC, Chen F, Zhao JN. Radiological model based on the standard magnetic resonance sequences for detecting methylguanine methyltransferase methylation in glioma using texture analysis. Cancer Sci 2021; 112:2835-2844. [PMID: 33932065 PMCID: PMC8253278 DOI: 10.1111/cas.14918] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 04/05/2021] [Accepted: 04/08/2021] [Indexed: 12/26/2022] Open
Abstract
This study aims to build a radiological model based on standard MR sequences for detecting methylguanine methyltransferase (MGMT) methylation in gliomas using texture analysis. A retrospective cross‐sectional study was undertaken in a cohort of 53 glioma patients who underwent standard preoperative magnetic resonance (MR) imaging. Conventional visual radiographic features and clinical factors were compared between MGMT promoter methylated and unmethylated groups. Texture analysis extracted the top five most powerful texture features of MR images in each sequence quantitatively for detecting the MGMT promoter methylation status. The radiomic signature (Radscore) was generated by a linear combination of the five features and estimates in each sequence. The combined model based on each Radscore was established using multivariate logistic regression analysis. A receiver operating characteristic (ROC) curve, nomogram, calibration, and decision curve analysis (DCA) were used to evaluate the performance of the model. No significant differences were observed in any of the visual radiographic features or clinical factors between different MGMT methylated statuses. The top five most powerful features were selected from a total of 396 texture features of T1, contrast‐enhanced T1, T2, and T2 FLAIR. Each sequence’s Radscore can distinguish MGMT methylated status. A combined model based on Radscores showed differentiation between methylated MGMT and unmethylated MGMT both in the glioblastoma (GBM) dataset as well as the dataset for all other gliomas. The area under the ROC curve values for the combined model was 0.818, with 90.5% sensitivity and 72.7% specificity, in the GBM dataset, and 0.833, with 70.2% sensitivity and 90.6% specificity, in the overall gliomas dataset. Nomogram, calibration, and DCA also validated the performance of the combined model. The combined model based on texture features could be considered as a noninvasive imaging marker for detecting MGMT methylation status in glioma.
Collapse
Affiliation(s)
- Wei-Yuan Huang
- Department of Radiology, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China.,Department of Radiology, Weill Medical College of Cornell University, New York, NY, USA
| | - Ling-Hua Wen
- Department of Radiology, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| | - Gang Wu
- Department of Radiotherapy, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| | - Pei-Pei Pang
- Department of Pharmaceuticals Diagnosis, GE Healthcare, Hangzhou, China
| | - Richard Ogbuji
- Department of Neurosurgery, The Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chao-Cai Zhang
- Department of Neurosurgery, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| | - Feng Chen
- Department of Radiology, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| | - Jian-Nong Zhao
- Department of Neurosurgery, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| |
Collapse
|
35
|
Jian A, Jang K, Manuguerra M, Liu S, Magnussen J, Di Ieva A. Machine Learning for the Prediction of Molecular Markers in Glioma on Magnetic Resonance Imaging: A Systematic Review and Meta-Analysis. Neurosurgery 2021; 89:31-44. [PMID: 33826716 DOI: 10.1093/neuros/nyab103] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 01/24/2021] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Molecular characterization of glioma has implications for prognosis, treatment planning, and prediction of treatment response. Current histopathology is limited by intratumoral heterogeneity and variability in detection methods. Advances in computational techniques have led to interest in mining quantitative imaging features to noninvasively detect genetic mutations. OBJECTIVE To evaluate the diagnostic accuracy of machine learning (ML) models in molecular subtyping gliomas on preoperative magnetic resonance imaging (MRI). METHODS A systematic search was performed following PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analysis) guidelines to identify studies up to April 1, 2020. Methodological quality of studies was assessed using the Quality Assessment for Diagnostic Accuracy Studies (QUADAS)-2. Diagnostic performance estimates were obtained using a bivariate model and heterogeneity was explored using metaregression. RESULTS Forty-four original articles were included. The pooled sensitivity and specificity for predicting isocitrate dehydrogenase (IDH) mutation in training datasets were 0.88 (95% CI 0.83-0.91) and 0.86 (95% CI 0.79-0.91), respectively, and 0.83 to 0.85 in validation sets. Use of data augmentation and MRI sequence type were weakly associated with heterogeneity. Both O6-methylguanine-DNA methyltransferase (MGMT) gene promoter methylation and 1p/19q codeletion could be predicted with a pooled sensitivity and specificity between 0.76 and 0.83 in training datasets. CONCLUSION ML application to preoperative MRI demonstrated promising results for predicting IDH mutation, MGMT methylation, and 1p/19q codeletion in glioma. Optimized ML models could lead to a noninvasive, objective tool that captures molecular information important for clinical decision making. Future studies should use multicenter data, external validation and investigate clinical feasibility of ML models.
Collapse
Affiliation(s)
- Anne Jian
- Computational NeuroSurgery (CNS) Lab, Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia.,Melbourne Medical School, University of Melbourne, Melbourne, Australia
| | - Kevin Jang
- Computational NeuroSurgery (CNS) Lab, Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia.,Discipline of Surgery, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Maurizio Manuguerra
- Department of Mathematics and Statistics, Faculty of Science and Engineering, Macquarie University, Sydney, Australia
| | - Sidong Liu
- Computational NeuroSurgery (CNS) Lab, Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia.,Centre for Health Informatics, Macquarie University, Sydney, Australia
| | - John Magnussen
- Computational NeuroSurgery (CNS) Lab, Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia.,Macquarie Medical Imaging, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
| | - Antonio Di Ieva
- Computational NeuroSurgery (CNS) Lab, Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia.,Macquarie Neurosurgery, Macquarie University, Sydney, Australia
| |
Collapse
|
36
|
Luo H, Zhuang Q, Wang Y, Abudumijiti A, Shi K, Rominger A, Chen H, Yang Z, Tran V, Wu G, Li Z, Fan Z, Qi Z, Guo Y, Yu J, Shi Z. A novel image signature-based radiomics method to achieve precise diagnosis and prognostic stratification of gliomas. J Transl Med 2021; 101:450-462. [PMID: 32829381 DOI: 10.1038/s41374-020-0472-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 07/14/2020] [Accepted: 07/15/2020] [Indexed: 12/23/2022] Open
Abstract
Radiomics has potential advantages in the noninvasive histopathological and molecular diagnosis of gliomas. We aimed to develop a novel image signature (IS)-based radiomics model to achieve multilayered preoperative diagnosis and prognostic stratification of gliomas. Herein, we established three separate case cohorts, consisting of 655 glioma patients, and carried out a retrospective study. Image and clinical data of three cohorts were used for training (N = 188), cross-validation (N = 411), and independent testing (N = 56) of the IS model. All tumors were segmented from magnetic resonance (MR) images by the 3D U-net, followed by extraction of high-throughput network features, which were referred to as IS. IS was then used to perform noninvasive histopathological diagnosis and molecular subtyping. Moreover, a new IS-based clustering method was applied for prognostic stratification in IDH-wild-type lower-grade glioma (IDHwt LGG) and triple-negative glioblastoma (1p19q retain/IDH wild-type/TERTp-wild-type GBM). The average accuracies of histological diagnosis and molecular subtyping were 89.8 and 86.1% in the cross-validation cohort, while these numbers reached 83.9 and 80.4% in the independent testing cohort. IS-based clustering method was demonstrated to successfully divide IDHwt LGG into two subgroups with distinct median overall survival time (48.63 vs 38.27 months respectively, P = 0.023), and two subgroups in triple-negative GBM with different median OS time (36.8 vs 18.2 months respectively, P = 0.013). Our findings demonstrate that our novel IS-based radiomics model is an effective tool to achieve noninvasive histo-molecular pathological diagnosis and prognostic stratification of gliomas. This IS model shows potential for future routine use in clinical practice.
Collapse
Affiliation(s)
- Huigao Luo
- Department of Electronic Engineering, Fudan University, Shanghai, China
| | - Qiyuan Zhuang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Yuanyuan Wang
- Department of Electronic Engineering, Fudan University, Shanghai, China
| | | | - Kuangyu Shi
- Department of Nuclear Medicine, University of Bern, Bern, Switzerland
- Department of Informatics, Technical University of Munich, Munich, Germany
| | - Axel Rominger
- Department of Nuclear Medicine, University of Bern, Bern, Switzerland
| | - Hong Chen
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhong Yang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Vanessa Tran
- B-BMed, The University of Melbourne, Melbourne, VIC, Australia
| | - Guoqing Wu
- Department of Electronic Engineering, Fudan University, Shanghai, China
| | - Zeju Li
- Department of Electronic Engineering, Fudan University, Shanghai, China
- Department of Computing, Imperial College, London, UK
| | - Zhen Fan
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Zengxin Qi
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Yuxiao Guo
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Jinhua Yu
- Department of Electronic Engineering, Fudan University, Shanghai, China.
- AI Lab of Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200433, China.
- Key Laboratory of Medical Imaging, Computing and Computer-Assisted Intervention, Shanghai, China.
| | - Zhifeng Shi
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
37
|
Le NQK, Hung TNK, Do DT, Lam LHT, Dang LH, Huynh TT. Radiomics-based machine learning model for efficiently classifying transcriptome subtypes in glioblastoma patients from MRI. Comput Biol Med 2021; 132:104320. [PMID: 33735760 DOI: 10.1016/j.compbiomed.2021.104320] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 03/05/2021] [Accepted: 03/05/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND In the field of glioma, transcriptome subtypes have been considered as an important diagnostic and prognostic biomarker that may help improve the treatment efficacy. However, existing identification methods of transcriptome subtypes are limited due to the relatively long detection period, the unattainability of tumor specimens via biopsy or surgery, and the fleeting nature of intralesional heterogeneity. In search of a superior model over previous ones, this study evaluated the efficiency of eXtreme Gradient Boosting (XGBoost)-based radiomics model to classify transcriptome subtypes in glioblastoma patients. METHODS This retrospective study retrieved patients from TCGA-GBM and IvyGAP cohorts with pathologically diagnosed glioblastoma, and separated them into different transcriptome subtypes groups. GBM patients were then segmented into three different regions of MRI: enhancement of the tumor core (ET), non-enhancing portion of the tumor core (NET), and peritumoral edema (ED). We subsequently used handcrafted radiomics features (n = 704) from multimodality MRI and two-level feature selection techniques (Spearman correlation and F-score tests) in order to find the features that could be relevant. RESULTS After the feature selection approach, we identified 13 radiomics features that were the most meaningful ones that can be used to reach the optimal results. With these features, our XGBoost model reached the predictive accuracies of 70.9%, 73.3%, 88.4%, and 88.4% for classical, mesenchymal, neural, and proneural subtypes, respectively. Our model performance has been improved in comparison with the other models as well as previous works on the same dataset. CONCLUSION The use of XGBoost and two-level feature selection analysis (Spearman correlation and F-score) could be expected as a potential combination for classifying transcriptome subtypes with high performance and might raise public attention for further research on radiomics-based GBM models.
Collapse
Affiliation(s)
- Nguyen Quoc Khanh Le
- Professional Master Program in Artificial Intelligence in Medicine, College of Medicine, Taipei Medical University, Taipei, 106, Taiwan; Research Center for Artificial Intelligence in Medicine, Taipei Medical University, Taipei, 106, Taiwan; Translational Imaging Research Center, Taipei Medical University Hospital, Taipei, 110, Taiwan.
| | - Truong Nguyen Khanh Hung
- International Master/Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan; Orthopedic and Trauma Department, Cho Ray Hospital, Ho Chi Minh City, 70000, Viet Nam
| | - Duyen Thi Do
- Graduate Institute of Biomedical Informatics, Taipei Medical University, Taipei, 106, Taiwan
| | - Luu Ho Thanh Lam
- International Master/Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan; Children's Hospital 2, Ho Chi Minh City, 70000, Viet Nam
| | - Luong Huu Dang
- Department of Otolaryngology, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, 70000, Viet Nam
| | - Tuan-Tu Huynh
- Department of Electrical Engineering, Yuan Ze University, No. 135, Yuandong Road, Zhongli, 320, Taoyuan, Taiwan; Department of Electrical Electronic and Mechanical Engineering, Lac Hong University, No. 10, Huynh Van Nghe Road, Bien Hoa, Dong Nai, 76120, Viet Nam
| |
Collapse
|
38
|
Comprehensive classification models based on amygdala radiomic features for Alzheimer's disease and mild cognitive impairment. Brain Imaging Behav 2021; 15:2377-2386. [PMID: 33537928 DOI: 10.1007/s11682-020-00434-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 11/21/2020] [Accepted: 12/17/2020] [Indexed: 11/26/2022]
Abstract
The amygdala is an important part of the medial temporal lobe and plays a pivotal role in the emotional and cognitive function. The aim of this study was to build and validate comprehensive classification models based on amygdala radiomic features for Alzheimer's disease (AD) and amnestic mild cognitive impairment (aMCI). For the amygdala, 3360 radiomic features were extracted from 97 AD patients, 53 aMCI patients and 45 normal controls (NCs) on the three-dimensional T1-weighted magnetization-prepared rapid gradient echo (MPRAGE) images. We used maximum relevance and minimum redundancy (mRMR) and least absolute shrinkage and selection operator (LASSO) to select the features. Multivariable logistic regression analysis was performed to build three classification models (AD-NC group, AD-aMCI group, and aMCI-NC group). Finally, internal validation was assessed. After two steps of feature selection, there were 5 radiomic features remained in the AD-NC group, 16 features remained in the AD-aMCI group and the aMCI-NC group, respectively. The proposed logistic classification analysis based on amygdala radiomic features achieves an accuracy of 0.90 and an area under the ROC curve (AUC) of 0.93 for AD vs. NC classification, an accuracy of 0.81 and an AUC of 0.84 for AD vs. aMCI classification, and an accuracy of 0.75 and an AUC of 0.80 for aMCI vs. NC classification. Amygdala radiomic features might be early biomarkers for detecting microstructural brain tissue changes during the AD and aMCI course. Logistic classification analysis demonstrated the promising classification performances for clinical applications among AD, aMCI and NC groups.
Collapse
|
39
|
Kong Z, Jiang C, Zhang Y, Liu S, Liu D, Liu Z, Chen W, Liu P, Yang T, Lyu Y, Zhao D, You H, Wang Y, Ma W, Feng F. Thin-Slice Magnetic Resonance Imaging-Based Radiomics Signature Predicts Chromosomal 1p/19q Co-deletion Status in Grade II and III Gliomas. Front Neurol 2020; 11:551771. [PMID: 33192984 PMCID: PMC7642873 DOI: 10.3389/fneur.2020.551771] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 09/23/2020] [Indexed: 12/13/2022] Open
Abstract
Objective: Chromosomal 1p/19q co-deletion is recognized as a diagnostic, prognostic, and predictive biomarker in lower grade glioma (LGG). This study aims to construct a radiomics signature to non-invasively predict the 1p/19q co-deletion status in LGG. Methods: Ninety-six patients with pathology-confirmed LGG were retrospectively included and randomly assigned into training (n = 78) and validation (n = 18) dataset. Three-dimensional contrast-enhanced T1 (3D-CE-T1)-weighted magnetic resonance (MR) images and T2-weighted MR images were acquired, and simulated-conventional contrast-enhanced T1 (SC-CE-T1)-weighted images were generated. One hundred and seven shape, first-order, and texture radiomics features were extracted from each imaging modality and selected using the least absolute shrinkage and selection operator on the training dataset. A 3D-radiomics signature based on 3D-CE-T1 and T2-weighted features and a simulated-conventional (SC) radiomics signature based on SC-CE-T1 and T2-weighted features were established using random forest. The radiomics signatures were validated independently and evaluated using receiver operating characteristic (ROC) curves. Tumors with IDH mutations were also separately assessed. Results: Four radiomics features were selected to construct the 3D-radiomics signature and displayed accuracies of 0.897 and 0.833, areas under the ROC curves (AUCs) of 0.940 and 0.889 in the training and validation datasets, respectively. The SC-radiomics signature was constructed with 4 features, but the AUC values were lower than that of the 3D signature. In the IDH-mutated subgroup, the 3D-radiomics signature presented AUCs of 0.950–1.000. Conclusions: The MRI-based radiomics signature can differentiate 1p/19q co-deletion status in LGG with or without predetermined IDH status. 3D-CE-T1-weighted radiomics features are more favorable than SC-CE-T1-weighted features in the establishment of radiomics signatures.
Collapse
Affiliation(s)
- Ziren Kong
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chendan Jiang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yiwei Zhang
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Sirui Liu
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Delin Liu
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zeyu Liu
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wenlin Chen
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Penghao Liu
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tianrui Yang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuelei Lyu
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Department of Radiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Dachun Zhao
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hui You
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yu Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wenbin Ma
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Feng Feng
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
40
|
Le NQK, Do DT, Chiu FY, Yapp EKY, Yeh HY, Chen CY. XGBoost Improves Classification of MGMT Promoter Methylation Status in IDH1 Wildtype Glioblastoma. J Pers Med 2020; 10:jpm10030128. [PMID: 32942564 PMCID: PMC7563334 DOI: 10.3390/jpm10030128] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 09/03/2020] [Accepted: 09/09/2020] [Indexed: 02/07/2023] Open
Abstract
Approximately 96% of patients with glioblastomas (GBM) have IDH1 wildtype GBMs, characterized by extremely poor prognosis, partly due to resistance to standard temozolomide treatment. O6-Methylguanine-DNA methyltransferase (MGMT) promoter methylation status is a crucial prognostic biomarker for alkylating chemotherapy resistance in patients with GBM. However, MGMT methylation status identification methods, where the tumor tissue is often undersampled, are time consuming and expensive. Currently, presurgical noninvasive imaging methods are used to identify biomarkers to predict MGMT methylation status. We evaluated a novel radiomics-based eXtreme Gradient Boosting (XGBoost) model to identify MGMT promoter methylation status in patients with IDH1 wildtype GBM. This retrospective study enrolled 53 patients with pathologically proven GBM and tested MGMT methylation and IDH1 status. Radiomics features were extracted from multimodality MRI and tested by F-score analysis to identify important features to improve our model. We identified nine radiomics features that reached an area under the curve of 0.896, which outperformed other classifiers reported previously. These features could be important biomarkers for identifying MGMT methylation status in IDH1 wildtype GBM. The combination of radiomics feature extraction and F-core feature selection significantly improved the performance of the XGBoost model, which may have implications for patient stratification and therapeutic strategy in GBM.
Collapse
Affiliation(s)
- Nguyen Quoc Khanh Le
- Professional Master Program in Artificial Intelligence in Medicine, College of Medicine, Taipei Medical University, Taipei City 106, Taiwan
- Research Center for Artificial Intelligence in Medicine, Taipei Medical University, Taipei City 106, Taiwan;
- Correspondence: (N.Q.K.L.); (C.-Y.C.); Tel.: +886-266-382-736 (ext. 1992) (N.Q.K.L.); Fax: +886-2-2732-1956 (N.Q.K.L.)
| | - Duyen Thi Do
- Faculty of Applied Sciences, Ton Duc Thang University, Ho Chi Minh City 70000, Vietnam;
| | - Fang-Ying Chiu
- Research Center for Artificial Intelligence in Medicine, Taipei Medical University, Taipei City 106, Taiwan;
| | - Edward Kien Yee Yapp
- Singapore Institute of Manufacturing Technology, 2 Fusionopolis Way, #08-04, Innovis, Singapore 138634, Singapore;
| | - Hui-Yuan Yeh
- Medical Humanities Research Cluster, School of Humanities, Nanyang Technological University, 48 Nanyang Ave, Singapore 639798, Singapore;
| | - Cheng-Yu Chen
- Professional Master Program in Artificial Intelligence in Medicine, College of Medicine, Taipei Medical University, Taipei City 106, Taiwan
- Research Center for Artificial Intelligence in Medicine, Taipei Medical University, Taipei City 106, Taiwan;
- Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Department of Medical Imaging, Taipei Medical University Hospital, Taipei 11031, Taiwan
- Correspondence: (N.Q.K.L.); (C.-Y.C.); Tel.: +886-266-382-736 (ext. 1992) (N.Q.K.L.); Fax: +886-2-2732-1956 (N.Q.K.L.)
| |
Collapse
|