1
|
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
|
2
|
He W, Huang W, Zhang L, Wu X, Zhang S, Zhang B. Radiogenomics: bridging the gap between imaging and genomics for precision oncology. MedComm (Beijing) 2024; 5:e722. [PMID: 39252824 PMCID: PMC11381657 DOI: 10.1002/mco2.722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 08/06/2024] [Accepted: 08/18/2024] [Indexed: 09/11/2024] Open
Abstract
Genomics allows the tracing of origin and evolution of cancer at molecular scale and underpin modern cancer diagnosis and treatment systems. Yet, molecular biomarker-guided clinical decision-making encounters major challenges in the realm of individualized medicine, consisting of the invasiveness of procedures and the sampling errors due to high tumor heterogeneity. By contrast, medical imaging enables noninvasive and global characterization of tumors at a low cost. In recent years, radiomics has overcomes the limitations of human visual evaluation by high-throughput quantitative analysis, enabling the comprehensive utilization of the vast amount of information underlying radiological images. The cross-scale integration of radiomics and genomics (hereafter radiogenomics) has the enormous potential to enhance cancer decoding and act as a catalyst for digital precision medicine. Herein, we provide a comprehensive overview of the current framework and potential clinical applications of radiogenomics in patient care. We also highlight recent research advances to illustrate how radiogenomics can address common clinical problems in solid tumors such as breast cancer, lung cancer, and glioma. Finally, we analyze existing literature to outline challenges and propose solutions, while also identifying future research pathways. We believe that the perspectives shared in this survey will provide a valuable guide for researchers in the realm of radiogenomics aiming to advance precision oncology.
Collapse
Affiliation(s)
- Wenle He
- Department of Radiology The First Affiliated Hospital of Jinan University Guangzhou Guangdong China
| | - Wenhui Huang
- Department of Radiology The First Affiliated Hospital of Jinan University Guangzhou Guangdong China
| | - Lu Zhang
- Department of Radiology The First Affiliated Hospital of Jinan University Guangzhou Guangdong China
| | - Xuewei Wu
- Department of Radiology The First Affiliated Hospital of Jinan University Guangzhou Guangdong China
| | - Shuixing Zhang
- Department of Radiology The First Affiliated Hospital of Jinan University Guangzhou Guangdong China
| | - Bin Zhang
- Department of Radiology The First Affiliated Hospital of Jinan University Guangzhou Guangdong China
| |
Collapse
|
3
|
Huang D, Gao T, Zhang Y, Lyu X, Liu S, Chen Y, Su C, Hu W, Lv Y. A Study on Prognosis of Diffuse Glioma Based on Clinical Factors and Magnetic Resonance Imaging Radiomics. World Neurosurg 2024; 186:e514-e530. [PMID: 38583562 DOI: 10.1016/j.wneu.2024.03.166] [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: 11/07/2023] [Revised: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 04/09/2024]
Abstract
OBJECTIVE To construct an optimal prognostic model to assess the prognosis of patients with diffuse glioma. METHODS Preoperative magnetic resonance imaging and clinical data were retrospectively collected from 266 patients (training cohort: validation cohort=7:3) with pathologically confirmed diffuse gliomas. A radiomics prognostic model (R-model) based on the radiomics features was constructed. A prognostic model based on clinical factors (C-model) and a fusion model (F-model) was also constructed. Based on the optimal model of three models, the nomogram was constructed. Finally, a "Prognosis Calculator for Diffuse Glioma" was constructed based on the nomogram. RESULTS The c-index of the R-, C-, and F-models in the validation cohort was 0.742, 0.796, and 0.814, respectively. In the validation cohort, the 1-year area under the curve of the R-, C-, and F-models was 0.749, 0.806, and 0.836, respectively; the 3-year area under the curve was 0.896, 0.966, and 0.963, respectively. In the training cohort, validation cohort, all cohorts, and different grades of glioma cohorts, F-model (optimal model) could identify low- and high-risk groups well. The "Prognosis Calculator for Diffuse Glioma" was available at https://github.com/HDCurry/prognosis. CONCLUSIONS Among the three models, the F-model (radiomics combined with clinical factors) had optimal predictive efficacy and could more accurately assess the prognosis of diffuse glioma. The "Prognosis Calculator for Diffuse Glioma" constructed based on this model could assist clinicians in more easily and accurately assessing the prognosis of patients with diffuse glioma, thus enabling them to make more reasonable treatment strategies.
Collapse
Affiliation(s)
- Dongcun Huang
- Department of Radiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Tianyu Gao
- Department of Radiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Ying Zhang
- Department of Radiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiaofei Lyu
- China Quality Certification Centre, Guangzhou, China
| | - Siheng Liu
- Department of Radiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yinsheng Chen
- Department of Neurosurgery/Neuro-Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Changliang Su
- Department of Radiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Wanming Hu
- Department of Pathology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yanchun Lv
- Department of Radiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China.
| |
Collapse
|
4
|
Park CJ, Kim S, Han K, Ahn SS, Kim D, Park YW, Chang JH, Kim SH, Lee SK. Diffusion- and Perfusion-Weighted MRI Radiomics for Survival Prediction in Patients with Lower-Grade Gliomas. Yonsei Med J 2024; 65:283-292. [PMID: 38653567 PMCID: PMC11045349 DOI: 10.3349/ymj.2023.0323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/27/2023] [Accepted: 12/13/2023] [Indexed: 04/25/2024] Open
Abstract
PURPOSE Lower-grade gliomas of histologic grades 2 and 3 follow heterogenous clinical outcomes, which necessitates risk stratification. This study aimed to evaluate whether diffusion-weighted and perfusion-weighted MRI radiomics allow overall survival (OS) prediction in patients with lower-grade gliomas and investigate its prognostic value. MATERIALS AND METHODS In this retrospective study, radiomic features were extracted from apparent diffusion coefficient, relative cerebral blood volume map, and Ktrans map in patients with pathologically confirmed lower-grade gliomas (January 2012-February 2019). The radiomics risk score (RRS) calculated from selected features constituted a radiomics model. Multivariable Cox regression analysis, including clinical features and RRS, was performed. The models' integrated area under the receiver operating characteristic curves (iAUCs) were compared. The radiomics model combined with clinical features was presented as a nomogram. RESULTS The study included 129 patients (median age, 44 years; interquartile range, 37-57 years; 63 female): 90 patients for training set and 39 patients for test set. The RRS was an independent risk factor for OS with a hazard ratio of 6.01. The combined clinical and radiomics model achieved superior performance for OS prediction compared to the clinical model in both training (iAUC, 0.82 vs. 0.72, p=0.002) and test sets (0.88 vs. 0.76, p=0.04). The radiomics nomogram combined with clinical features exhibited good agreement between the actual and predicted OS with C-index of 0.83 and 0.87 in the training and test sets, respectively. CONCLUSION Adding diffusion- and perfusion-weighted MRI radiomics to clinical features improved survival prediction in lower-grade glioma.
Collapse
Affiliation(s)
- Chae Jung Park
- Department of Radiology, Research Institute of Radiological Science, Yongin Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Sooyon Kim
- Department of Applied Statistics, Yonsei University, Seoul, Korea
| | - Kyunghwa Han
- Department of Radiology, Center for Clinical Imaging Data Science, Research Institute of Radiological Sciences, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Sung Soo Ahn
- Department of Radiology, Center for Clinical Imaging Data Science, Research Institute of Radiological Sciences, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.
| | - Dain Kim
- Graduate School of Artificial Intelligence, Pohang University of Science and Technology, Pohang, Korea
| | - Yae Won Park
- Department of Radiology, Center for Clinical Imaging Data Science, Research Institute of Radiological Sciences, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Jong Hee Chang
- Department of Neurosurgery, Yonsei University College of Medicine, Seoul, Korea
| | - Se Hoon Kim
- Department of Pathology, Yonsei University College of Medicine, Seoul, Korea
| | - Seung-Koo Lee
- Department of Radiology, Center for Clinical Imaging Data Science, Research Institute of Radiological Sciences, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
5
|
Li X, Cheng Y, Han X, Cui B, Li J, Yang H, Xu G, Lin Q, Xiao X, Tang J, Lu J. Exploring the association of glioma tumor residuals from incongruent [ 18F]FET PET/MR imaging with tumor proliferation using a multiparametric MRI radiomics nomogram. Eur J Nucl Med Mol Imaging 2024; 51:779-796. [PMID: 37864593 DOI: 10.1007/s00259-023-06468-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 09/28/2023] [Indexed: 10/23/2023]
Abstract
PURPOSE The study aimed to using multiparametric MRI radiomics to predict glioma tumor residuals (TRFET over MR) derived from incongruent [18F]fluoroethyl-L-tyrosine ([18F]FET) PET/MR imaging. METHODS One hundred ten patients with gliomas who underwent [18F]FET PET/MR scanning were retrospectively analyzed. The TRFET over MR was identified by the discrepancy-PET that the extent of resection (EOR) based on MRI subtracted the biological tumor volume on PET images. The MRI parameters and radiomics features were extracted based on EOR and selected by the least absolute shrinkage and selection operator to construct radiomics score (Rad-score). The correlation network analysis of all features was analyzed by Spearman's correlation tests. The methods for evaluating the clinical usefulness consisted of the receiver operating characteristic curve, the calibration curve, and decision curve analysis. RESULTS The Rad-score of the patients with the TRFET over MR was significantly higher than those with the non TRFET over MR (p < 0.001). The Rad-score was significantly correlated with the discrepancy-PET (r = 0.72, p < 0.001), Ki-67 level (r = 0.76, p < 0.001), and epidermal growth factor receptor (EGFR) of gliomas (r = 0.75, p < 0.001), respectively. Moreover, there was a difference of the correlation network analysis between the TRPET over MR group and non TRFET over MR group. The nomogram combing Rad-score and clinical features had the greatest performance in predicting TRFET over MR (AUC = 0.90/0.87, training/testing). There was a significant difference in prognosis (median OS, 17 m vs. 43 m) between patients with TRFET over MR and non TRFET over MR based on nomogram prediction (p < 0.001). CONCLUSION The nomogram based on MRI radiomics would predict gliomas tumor residuals caused by the absence of 18F-PET PET examination and adjust EOR to improve prognosis.
Collapse
Affiliation(s)
- Xiaoran Li
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Capital Medical University, Beijing, China
| | - Ye Cheng
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xin Han
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Capital Medical University, Beijing, China
| | - Bixiao Cui
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Capital Medical University, Beijing, China
| | - Jing Li
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Capital Medical University, Beijing, China
| | - Hongwei Yang
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Capital Medical University, Beijing, China
| | - Geng Xu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Qingtang Lin
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xinru Xiao
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jie Tang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China.
| | - Jie Lu
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China.
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Capital Medical University, Beijing, China.
| |
Collapse
|
6
|
Zhao Z, Song Z, Wang Z, Zhang F, Ding Z, Fan T. Advances in Molecular Pathology, Diagnosis and Treatment of Spinal Cord Astrocytomas. Technol Cancer Res Treat 2024; 23:15330338241262483. [PMID: 39043042 PMCID: PMC11271101 DOI: 10.1177/15330338241262483] [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: 02/17/2024] [Revised: 05/23/2024] [Accepted: 05/28/2024] [Indexed: 07/25/2024] Open
Abstract
Spinal cord astrocytoma (SCA) is a rare subtype of astrocytoma, posing challenges in diagnosis and treatment. Low-grade SCA can achieve long-term survival solely through surgery, while high-grade has a disappointing prognosis even with comprehensive treatment. Diagnostic criteria and standard treatment of intracranial astrocytoma have shown obvious limitations in SCA. Research on the molecular mechanism in SCA is lagging far behind that on intracranial astrocytoma. In recent years, huge breakthroughs have been made in molecular pathology of astrocytoma, and novel techniques have emerged, including DNA methylation analysis and radiomics. These advances are now making it possible to provide a precise diagnosis and develop corresponding treatment strategies in SCA. Our aim is to review the current status of diagnosis and treatment of SCA, and summarize the latest research advancement, including tumor subtype, molecular characteristics, diagnostic technology, and potential therapy strategies, thus deepening our understanding of this uncommon tumor type and providing guidance for accurate diagnosis and treatment.
Collapse
Affiliation(s)
- Zijun Zhao
- Spine Center, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Zihan Song
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Zairan Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Fan Zhang
- Spine Center, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Ze Ding
- Spine Center, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Tao Fan
- Spine Center, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
7
|
Chen C, Du X, Yang L, Liu H, Li Z, Gou Z, Qi J. Research on application of radiomics in glioma: a bibliometric and visual analysis. Front Oncol 2023; 13:1083080. [PMID: 37771434 PMCID: PMC10523166 DOI: 10.3389/fonc.2023.1083080] [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: 10/28/2022] [Accepted: 08/16/2023] [Indexed: 09/30/2023] Open
Abstract
Background With the continuous development of medical imaging informatics technology, radiomics has become a new and evolving field in medical applications. Radiomics aims to be an aid to support clinical decision making by extracting quantitative features from medical images and has a very wide range of applications. The purpose of this study was to perform a bibliometric and visual analysis of scientific results and research trends in the research application of radiomics in glioma. Methods We searched the Web of Science Core Collection (WOScc) for publications related to glioma radiomics. A bibliometric and visual analysis of online publications in this field related to countries/regions, authors, journals, references and keywords was performed using CiteSpace and R software. Results A total of 587 relevant literature published from 2012 to September 2022 were retrieved in WOScc, and finally a total of 484 publications were obtained according to the filtering criteria, including 393 (81.20%) articles and 91 (18.80%) reviews. The number of relevant publications increases year by year. The highest number of publications was from the USA (171 articles, 35.33%) and China (170 articles, 35.12%). The research institution with the highest number of publications was Chinese Acad Sci (24), followed by Univ Penn (22) and Fudan Univ (21). WANG Y (27) had the most publications, followed by LI Y (22), and WANG J (20). Among the 555 co-cited authors, LOUIS DN (207) and KICKINGEREDER P (207) were the most cited authors. FRONTIERS IN ONCOLOGY (42) was the most published journal and NEURO-ONCOLOGY (412) was the most co-cited journal. The most frequent keywords in all publications included glioblastoma (187), survival (136), classification (131), magnetic resonance imaging (113), machine learning (100), tumor (82), and feature (79), central nervous system (66), IDH (57), and radiomics (55). Cluster analysis was performed on the basis of keyword co-occurrence, and a total of 16 clusters were formed, indicating that these directions are the current hotspots of radiomics research applications in glioma and may be the future directions of continuous development. Conclusion In the past decade, radiomics has received much attention in the medical field and has been widely used in clinical research applications. Cooperation and communication between countries/regions need to be enhanced in future research to promote the development of radiomics in the field of medicine. In addition, the application of radiomics has improved the accuracy of pre-treatment diagnosis, efficacy prediction and prognosis assessment of glioma and helped to promote the development into precision medicine, the future still faces many challenges.
Collapse
Affiliation(s)
- Chunbao Chen
- Department of Neurosurgery, Afiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Xue Du
- Department of Oncology, The People's Hospital of Hechuan, Chongqing, China
- Department of Oncology, North Sichuan Medical College, Nanchong, China
| | - Lu Yang
- Department of Oncology, Suining Central Hospital, Suining, China
| | - Hongjun Liu
- Department of Neurosurgery, Afiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Zhou Li
- Department of Neurosurgery, Nanchong Central Hospital, The Afiliated Nanchong Central Hospital of North Sichuan Medical College, Nanchong, China
| | - Zhangyang Gou
- Department of Neurosurgery, Afiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Jian Qi
- Department of Neurosurgery, Afiliated Hospital of North Sichuan Medical College, Nanchong, China
| |
Collapse
|
8
|
Zhang H, Fan X, Zhang J, Wei Z, Feng W, Hu Y, Ni J, Yao F, Zhou G, Wan C, Zhang X, Wang J, Liu Y, You Y, Yu Y. Deep-learning and conventional radiomics to predict IDH genotyping status based on magnetic resonance imaging data in adult diffuse glioma. Front Oncol 2023; 13:1143688. [PMID: 37711207 PMCID: PMC10499353 DOI: 10.3389/fonc.2023.1143688] [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: 01/13/2023] [Accepted: 08/17/2023] [Indexed: 09/16/2023] Open
Abstract
Objectives In adult diffuse glioma, preoperative detection of isocitrate dehydrogenase (IDH) status helps clinicians develop surgical strategies and evaluate patient prognosis. Here, we aim to identify an optimal machine-learning model for prediction of IDH genotyping by combining deep-learning (DL) signatures and conventional radiomics (CR) features as model predictors. Methods In this study, a total of 486 patients with adult diffuse gliomas were retrospectively collected from our medical center (n=268) and the public database (TCGA, n=218). All included patients were randomly divided into the training and validation sets by using nested 10-fold cross-validation. A total of 6,736 CR features were extracted from four MRI modalities in each patient, namely T1WI, T1CE, T2WI, and FLAIR. The LASSO algorithm was performed for CR feature selection. In each MRI modality, we applied a CNN+LSTM-based neural network to extract DL features and integrate these features into a DL signature after the fully connected layer with sigmoid activation. Eight classic machine-learning models were analyzed and compared in terms of their prediction performance and stability in IDH genotyping by combining the LASSO-selected CR features and integrated DL signatures as model predictors. In the validation sets, the prediction performance was evaluated by using accuracy and the area under the curve (AUC) of the receiver operating characteristics, while the model stability was analyzed by using the relative standard deviation of the AUC (RSDAUC). Subgroup analyses of DL signatures and CR features were also individually conducted to explore their independent prediction values. Results Logistic regression (LR) achieved favorable prediction performance (AUC: 0.920 ± 0.043, accuracy: 0.843 ± 0.044), whereas support vector machine with the linear kernel (l-SVM) displayed low prediction performance (AUC: 0.812 ± 0.052, accuracy: 0.821 ± 0.050). With regard to stability, LR also showed high robustness against data perturbation (RSDAUC: 4.7%). Subgroup analyses showed that DL signatures outperformed CR features (DL, AUC: 0.915 ± 0.054, accuracy: 0.835 ± 0.061, RSDAUC: 5.9%; CR, AUC: 0.830 ± 0.066, accuracy: 0.771 ± 0.051, RSDAUC: 8.0%), while DL and DL+CR achieved similar prediction results. Conclusion In IDH genotyping, LR is a promising machine-learning classification model. Compared with CR features, DL signatures exhibit markedly superior prediction values and discriminative capability.
Collapse
Affiliation(s)
- Hongjian Zhang
- Department of Medical Informatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiao Fan
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Junxia Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhiyuan Wei
- Department of Medical Informatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wei Feng
- Department of Medical Informatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yifang Hu
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jiaying Ni
- Department of Medical Informatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Fushen Yao
- Department of Medical Informatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Gaoxin Zhou
- Department of Medical Informatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute of Medical Informatics and Management, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Cheng Wan
- Department of Medical Informatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute of Medical Informatics and Management, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xin Zhang
- Department of Medical Informatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute of Medical Informatics and Management, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Junjie Wang
- Department of Medical Informatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute of Medical Informatics and Management, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yun Liu
- Department of Medical Informatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute of Medical Informatics and Management, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yongping You
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yun Yu
- Department of Medical Informatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute of Medical Informatics and Management, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
9
|
Picca A, Bruno F, Nichelli L, Sanson M, Rudà R. Advances in molecular and imaging biomarkers in lower-grade gliomas. Expert Rev Neurother 2023; 23:1217-1231. [PMID: 37982735 DOI: 10.1080/14737175.2023.2285472] [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: 08/07/2023] [Accepted: 11/15/2023] [Indexed: 11/21/2023]
Abstract
INTRODUCTION Lower-grade (grade 2-3) gliomas (LGGs) constitutes a group of primary brain tumors with variable clinical behaviors and treatment responses. Recent advancements in molecular biology have redefined their classification, and novel imaging modalities emerged for the noninvasive diagnosis and follow-up. AREAS COVERED This review comprehensively analyses the current knowledge on molecular and imaging biomarkers in LGGs. Key molecular alterations, such as IDH mutations and 1p/19q codeletion, are discussed for their prognostic and predictive implications in guiding treatment decisions. Moreover, the authors explore theranostic biomarkers for the potential of tailored therapies. Additionally, they also describe the utility of advanced imaging modalities, including widely available techniques, as dynamic susceptibility contrast perfusion-weighted imaging and less validated, emerging approaches, for the noninvasive LGGs characterization and follow-up. EXPERT OPINION The integration of molecular markers enhanced the stratification of LGGs, leading to the new concept of integrated histomolecular classification. While the IDH mutation is an established key prognostic and predictive marker, recent results from IDH inhibitors trials showed its potential value as a theranostic marker. In this setting, advanced MRI techniques such as 2-D-hydroxyglutarate spectroscopy are very promising for the noninvasive diagnosis and monitoring of LGGs. This progress offers exciting prospects for personalized medicine and improved treatment outcomes in LGGs.
Collapse
Affiliation(s)
- Alberto Picca
- Service de Neurologie 2 Mazarin, Hôpital Universitaire Pitié-Salpêtrière, AP-HP, Paris, France
- Sorbonne Université, Inserm, CNRS, UMRS1127, Institut du Cerveau-Paris Brain Institute-ICM, AP-HP, Paris, France
| | - Francesco Bruno
- Division of Neuro-Oncology, Department of Neuroscience "Rita Levi Montalcini", University and City of Health and Science University Hospital, Turin, Italy
| | - Lucia Nichelli
- Service de Neuroradiologie, Hôpital Universitaire Pitié-Salpêtrière, AP-HP, Paris, France
| | - Marc Sanson
- Service de Neurologie 2 Mazarin, Hôpital Universitaire Pitié-Salpêtrière, AP-HP, Paris, France
- Sorbonne Université, Inserm, CNRS, UMRS1127, Institut du Cerveau-Paris Brain Institute-ICM, AP-HP, Paris, France
| | - Roberta Rudà
- Division of Neuro-Oncology, Department of Neuroscience "Rita Levi Montalcini", University and City of Health and Science University Hospital, Turin, Italy
| |
Collapse
|
10
|
Zheng J, Dong H, Li M, Lin X, Wang C. Prediction of IDH1 gene mutation by a nomogram based on multiparametric and multiregional MR images. Clinics (Sao Paulo) 2023; 78:100238. [PMID: 37354775 DOI: 10.1016/j.clinsp.2023.100238] [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: 02/22/2023] [Revised: 05/19/2023] [Accepted: 06/06/2023] [Indexed: 06/26/2023] Open
Abstract
OBJECTIVE To investigate the value of a nomogram based on multiparametric and multiregional MR images to predict Isocitrate Dehydrogenase-1 (IDH1) gene mutations in glioma. DATA AND METHODS The authors performed a retrospective analysis of 110 MR images of surgically confirmed pathological gliomas; 33 patients with IDH1 gene Mutation (IDH1-M) and 77 patients with Wild-type IDH1 (IDH1-W) were divided into training and validation sets in a 7:3 ratio. The clinical features were statistically analyzed using SPSS and R software. Three glioma regions (rCET, rE, rNEC) were outlined using ITK-SNAP software and projected to four conventional sequences (T1, T2, Flair, T1C) for feature extraction using AI-Kit software. The extracted features were screened using R software. A logistic regression model was established, and a nomogram was generated using the selected clinical features. Eight models were developed based on different sequences and ROIs, and Receiver Operating Characteristic (ROC) curves were used to evaluate the predictive efficacy. Decision curve analysis was performed to assess the clinical usefulness. RESULTS Age was selected with Radscore to construct the nomogram. The Model 1 AUC values based on four sequences and three ROIs were the highest in these models, at 0.93 and 0.89, respectively. Decision curve analysis indicated that the net benefit of model 1 was higher than that of the other models for most Pt-values. CONCLUSION A nomogram based on multiparametric and multiregional MR images can predict the mutation status of the IDH1 gene accurately.
Collapse
Affiliation(s)
- Jinjing Zheng
- Department of Radiology, Ningbo Medical Center Lihuili Hospital, Ningbo University, China
| | - Haibo Dong
- Department of Radiology, Ningbo Medical Center Lihuili Hospital, Ningbo University, China.
| | - Ming Li
- Department of Radiology, Ningbo Medical Center Lihuili Hospital, Ningbo University, China
| | - Xueyao Lin
- Department of Radiology, Ningbo Medical Center Lihuili Hospital, Ningbo University, China
| | - Chaochao Wang
- Department of Radiology, Ningbo Medical Center Lihuili Hospital, Ningbo University, China
| |
Collapse
|
11
|
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
|
12
|
Salome P, Sforazzini F, Grugnara G, Kudak A, Dostal M, Herold-Mende C, Heiland S, Debus J, Abdollahi A, Knoll M. MR Intensity Normalization Methods Impact Sequence Specific Radiomics Prognostic Model Performance in Primary and Recurrent High-Grade Glioma. Cancers (Basel) 2023; 15:cancers15030965. [PMID: 36765922 PMCID: PMC9913466 DOI: 10.3390/cancers15030965] [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: 01/07/2023] [Revised: 01/30/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
PURPOSE This study investigates the impact of different intensity normalization (IN) methods on the overall survival (OS) radiomics models' performance of MR sequences in primary (pHGG) and recurrent high-grade glioma (rHGG). METHODS MR scans acquired before radiotherapy were retrieved from two independent cohorts (rHGG C1: 197, pHGG C2: 141) from multiple scanners (15, 14). The sequences are T1 weighted (w), contrast-enhanced T1w (T1wce), T2w, and T2w-FLAIR. Sequence-specific significant features (SF) associated with OS, extracted from the tumour volume, were derived after applying 15 different IN methods. Survival analyses were conducted using Cox proportional hazard (CPH) and Poisson regression (POI) models. A ranking score was assigned based on the 10-fold cross-validated (CV) concordance index (C-I), mean square error (MSE), and the Akaike information criterion (AICs), to evaluate the methods' performance. RESULTS Scatter plots of the 10-CV C-I and MSE against the AIC showed an impact on the survival predictions between the IN methods and MR sequences (C1/C2 C-I range: 0.62-0.71/0.61-0.72, MSE range: 0.20-0.42/0.13-0.22). White stripe showed stable results for T1wce (C1/C2 C-I: 0.71/0.65, MSE: 0.21/0.14). Combat (0.68/0.62, 0.22/0.15) and histogram matching (HM, 0.67/0.64, 0.22/0.15) showed consistent prediction results for T2w models. They were also the top-performing methods for T1w in C2 (Combat: 0.67, 0.13; HM: 0.67, 0.13); however, only HM achieved high predictions in C1 (0.66, 0.22). After eliminating IN impacted SF using Spearman's rank-order correlation coefficient, a mean decrease in the C-I and MSE of 0.05 and 0.03 was observed in all four sequences. CONCLUSION The IN method impacted the predictive power of survival models; thus, performance is sequence-dependent.
Collapse
Affiliation(s)
- Patrick Salome
- Clinical Cooperation Unit (CCU) Radiation Oncology, German Cancer Research Centre, INF 280, 69120 Heidelberg, Germany
- Heidelberg Medical Faculty, Heidelberg University, 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK) Core Centre Heidelberg, 69120 Heidelberg, Germany
- Heidelberg Ion-Beam Therapy Centre (HIT), INF 450, 69120 Heidelberg, Germany
- Correspondence: (P.S.); (M.K.)
| | - Francesco Sforazzini
- Clinical Cooperation Unit (CCU) Radiation Oncology, German Cancer Research Centre, INF 280, 69120 Heidelberg, Germany
- Heidelberg Medical Faculty, Heidelberg University, 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK) Core Centre Heidelberg, 69120 Heidelberg, Germany
| | - Gianluca Grugnara
- Department of Neuroradiology, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Andreas Kudak
- Heidelberg Ion-Beam Therapy Centre (HIT), INF 450, 69120 Heidelberg, Germany
- Department of Radiation Oncology, Heidelberg University Hospital, INF 400, 69120 Heidelberg, Germany
- CCU Radiation Therapy, German Cancer Research Centre, INF 280, 69120 Heidelberg, Germany
| | - Matthias Dostal
- Heidelberg Ion-Beam Therapy Centre (HIT), INF 450, 69120 Heidelberg, Germany
- Department of Radiation Oncology, Heidelberg University Hospital, INF 400, 69120 Heidelberg, Germany
- CCU Radiation Therapy, German Cancer Research Centre, INF 280, 69120 Heidelberg, Germany
| | - Christel Herold-Mende
- Brain Tumour Group, European Organization for Research and Treatment of Cancer, 1200 Brussels, Belgium
- Division of Neurosurgical Research, Department of Neurosurgery, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Sabine Heiland
- Department of Neuroradiology, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Jürgen Debus
- German Cancer Consortium (DKTK) Core Centre Heidelberg, 69120 Heidelberg, Germany
- Heidelberg Ion-Beam Therapy Centre (HIT), INF 450, 69120 Heidelberg, Germany
- Department of Radiation Oncology, Heidelberg University Hospital, INF 400, 69120 Heidelberg, Germany
| | - Amir Abdollahi
- Clinical Cooperation Unit (CCU) Radiation Oncology, German Cancer Research Centre, INF 280, 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK) Core Centre Heidelberg, 69120 Heidelberg, Germany
- Heidelberg Ion-Beam Therapy Centre (HIT), INF 450, 69120 Heidelberg, Germany
- Department of Radiation Oncology, Heidelberg University Hospital, INF 400, 69120 Heidelberg, Germany
| | - Maximilian Knoll
- Clinical Cooperation Unit (CCU) Radiation Oncology, German Cancer Research Centre, INF 280, 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK) Core Centre Heidelberg, 69120 Heidelberg, Germany
- Heidelberg Ion-Beam Therapy Centre (HIT), INF 450, 69120 Heidelberg, Germany
- Department of Radiation Oncology, Heidelberg University Hospital, INF 400, 69120 Heidelberg, Germany
- Correspondence: (P.S.); (M.K.)
| |
Collapse
|
13
|
Zhai Y, Bai J, Xue Y, Li M, Mao W, Zhang X, Zhang Y. Development and validation of a preoperative MRI-based radiomics nomogram to predict progression-free survival in patients with clival chordomas. Front Oncol 2022; 12:996262. [PMID: 36591445 PMCID: PMC9800789 DOI: 10.3389/fonc.2022.996262] [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: 07/17/2022] [Accepted: 12/01/2022] [Indexed: 12/23/2022] Open
Abstract
Objectives The aim of this study was to establish and validate a MRI-based radiomics nomogram to predict progression-free survival (PFS) of clival chordoma. Methods A total of 174 patients were enrolled in the study (train cohort: 121 cases, test cohort: 53 cases). Radiomic features were extracted from multiparametric MRIs. Intraclass correlation coefficient analysis and a Lasso and Elastic-Net regularized generalized linear model were used for feature selection. Then, a nomogram was established via univariate and multivariate Cox regression analysis in the train cohort. The performance of this nomogram was assessed by area under curve (AUC) and calibration curve. Results A total of 3318 radiomic features were extracted from each patient, of which 2563 radiomic features were stable features. After feature selection, seven radiomic features were selected. Cox regression analysis revealed that 2 clinical factors (degree of resection, and presence or absence of primary chordoma) and 4 radiomic features were independent prognostic factors. The AUC of the established nomogram was 0.747, 0.807, and 0.904 for PFS prediction at 1, 3, and 5 years in the train cohort, respectively, compared with 0.582, 0.852, and 0.914 in the test cohort. Calibration and risk score stratified survival curves were satisfactory in the train and test cohort. Conclusions The presented nomogram demonstrated a favorable predictive accuracy of PFS, which provided a novel tool to predict prognosis and risk stratification. Our results suggest that radiomic analysis can effectively help neurosurgeons perform individualized evaluations of patients with clival chordomas.
Collapse
Affiliation(s)
- Yixuan Zhai
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Jiwei Bai
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Yake Xue
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mingxuan Li
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Wenbin Mao
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xuezhi Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yazhuo Zhang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China,Center of Brain Tumor, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China,China National Clinical Research Center for Neurological Diseases, Beijing, China,*Correspondence: Yazhuo Zhang,
| |
Collapse
|
14
|
Wang H, Zhang S, Xing X, Yue Q, Feng W, Chen S, Zhang J, Xie D, Chen N, Liu Y. Radiomic study on preoperative multi-modal magnetic resonance images identifies IDH-mutant TERT promoter-mutant gliomas. Cancer Med 2022; 12:2524-2537. [PMID: 36176070 PMCID: PMC9939206 DOI: 10.1002/cam4.5097] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 06/20/2022] [Accepted: 07/13/2022] [Indexed: 11/09/2022] Open
Abstract
OBJECTIVES Gliomas with comutations of isocitrate dehydrogenase (IDH) genes and telomerase reverse transcriptase (TERT) gene promoter (IDHmut pTERTmut) show distinct biological features and respond to first-line treatment differently in comparison with other gliomas. This study aimed to characterize the IDHmut pTERTmut gliomas in multimodal MRI using the radiomic method and establish a precise diagnostic model identifying this group of gliomas. METHODS A total of 140 patients with untreated primary gliomas were admitted between 2016 and 2020 to West China Hospital as a discovery cohort, including 22 IDHmut pTERTmut patients. Thirty-four additional cases from a different hospital were included in the study as an independent validation cohort. A total of 3654 radiomic features were extracted from the preoperative multimodal MRI images (T1c, FLAIR, and ADC maps) and filtered in a data-driven approach. The discovery cohort was split into training and test sets by a 4:1 ratio. A diagnostic model (multilayer perceptron classifier) for detecting the IDHmut pTERTmut gliomas was trained using an automatic machine-learning algorithm named tree-based pipeline optimization tool (TPOT). The most critical radiomic features in the model were identified and visualized. RESULTS The model achieved an area under the receiver-operating curve (AUROC) of 0.971 (95% CI, 0.902-1.000), the sensitivity of 0.833 (95% CI, 0.333-1.000), and the specificity of 0.966 (95% CI, 0.931-1.000) in the test set. The area under the precision-recall curve (AUCPR) was 0.754 (95% CI, 0.572-0.833) and the F1 score was 0.833 (95% CI, 0.500-1.000). In the independent validation set, the model reached 0.952 AUROC, 0.714 sensitivity, 0.963 specificity, 0.841 AUCPR, and 0.769 F1 score. MR radiomic features of the IDHmut pTERTmut gliomas represented homogenous low-complexity texture in three modalities. CONCLUSIONS An accurate diagnostic model was constructed for detecting IDHmut pTERTmut gliomas using multimodal radiomic features. The most important features were associated with the homogenous simple texture of IDHmut pTERTmut gliomas in MRI images transformed using Laplacian of Gaussian and wavelet filters.
Collapse
Affiliation(s)
- Haoyu Wang
- Department of NeurosurgeryWest China Hospital of Sichuan UniversityChengduChina,Department of NeurosurgeryXinhua Hospital, Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Shuxin Zhang
- Department of NeurosurgeryWest China Hospital of Sichuan UniversityChengduChina,Department of Head and Neck Surgery, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of MedicineUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Xiang Xing
- Department of NeurosurgeryWest China Hospital of Sichuan UniversityChengduChina
| | - Qiang Yue
- Department of RadiologyWest China Hospital of Sichuan UniversityChengduChina
| | - Wentao Feng
- Department of NeurosurgeryWest China Hospital of Sichuan UniversityChengduChina
| | - Siliang Chen
- Department of NeurosurgeryWest China Hospital of Sichuan UniversityChengduChina
| | - Jun Zhang
- Frontier Science Center for Disease Molecular Network, State Key Laboratory of BiotherapyWest China Hospital of Sichuan UniversityChengduChina
| | - Dan Xie
- Frontier Science Center for Disease Molecular Network, State Key Laboratory of BiotherapyWest China Hospital of Sichuan UniversityChengduChina
| | - Ni Chen
- Department of Pathology, West China HospitalSichuan UniversityChengduSichuanChina
| | - Yanhui Liu
- Department of NeurosurgeryWest China Hospital of Sichuan UniversityChengduChina
| |
Collapse
|
15
|
di Noia C, Grist JT, Riemer F, Lyasheva M, Fabozzi M, Castelli M, Lodi R, Tonon C, Rundo L, Zaccagna F. Predicting Survival in Patients with Brain Tumors: Current State-of-the-Art of AI Methods Applied to MRI. Diagnostics (Basel) 2022; 12:diagnostics12092125. [PMID: 36140526 PMCID: PMC9497964 DOI: 10.3390/diagnostics12092125] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/05/2022] [Accepted: 08/17/2022] [Indexed: 11/24/2022] Open
Abstract
Given growing clinical needs, in recent years Artificial Intelligence (AI) techniques have increasingly been used to define the best approaches for survival assessment and prediction in patients with brain tumors. Advances in computational resources, and the collection of (mainly) public databases, have promoted this rapid development. This narrative review of the current state-of-the-art aimed to survey current applications of AI in predicting survival in patients with brain tumors, with a focus on Magnetic Resonance Imaging (MRI). An extensive search was performed on PubMed and Google Scholar using a Boolean research query based on MeSH terms and restricting the search to the period between 2012 and 2022. Fifty studies were selected, mainly based on Machine Learning (ML), Deep Learning (DL), radiomics-based methods, and methods that exploit traditional imaging techniques for survival assessment. In addition, we focused on two distinct tasks related to survival assessment: the first on the classification of subjects into survival classes (short and long-term or eventually short, mid and long-term) to stratify patients in distinct groups. The second focused on quantification, in days or months, of the individual survival interval. Our survey showed excellent state-of-the-art methods for the first, with accuracy up to ∼98%. The latter task appears to be the most challenging, but state-of-the-art techniques showed promising results, albeit with limitations, with C-Index up to ∼0.91. In conclusion, according to the specific task, the available computational methods perform differently, and the choice of the best one to use is non-univocal and dependent on many aspects. Unequivocally, the use of features derived from quantitative imaging has been shown to be advantageous for AI applications, including survival prediction. This evidence from the literature motivates further research in the field of AI-powered methods for survival prediction in patients with brain tumors, in particular, using the wealth of information provided by quantitative MRI techniques.
Collapse
Affiliation(s)
- Christian di Noia
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum—University of Bologna, 40125 Bologna, Italy
| | - James T. Grist
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford OX1 3PT, UK
- Department of Radiology, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK
- Oxford Centre for Clinical Magnetic Research Imaging, University of Oxford, Oxford OX3 9DU, UK
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham B15 2SY, UK
| | - Frank Riemer
- Mohn Medical Imaging and Visualization Centre (MMIV), Department of Radiology, Haukeland University Hospital, N-5021 Bergen, Norway
| | - Maria Lyasheva
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Miriana Fabozzi
- Centro Medico Polispecialistico (CMO), 80058 Torre Annunziata, Italy
| | - Mauro Castelli
- NOVA Information Management School (NOVA IMS), Universidade NOVA de Lisboa, Campus de Campolide, 1070-312 Lisboa, Portugal
| | - Raffaele Lodi
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum—University of Bologna, 40125 Bologna, Italy
- Functional and Molecular Neuroimaging Unit, IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy
| | - Caterina Tonon
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum—University of Bologna, 40125 Bologna, Italy
- Functional and Molecular Neuroimaging Unit, IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy
| | - Leonardo Rundo
- Department of Information and Electrical Engineering and Applied Mathematics, University of Salerno, 84084 Fisciano, Italy
| | - Fulvio Zaccagna
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum—University of Bologna, 40125 Bologna, Italy
- Functional and Molecular Neuroimaging Unit, IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy
- Correspondence: ; Tel.: +39-0514969951
| |
Collapse
|
16
|
Xu C, Peng Y, Zhu W, Chen Z, Li J, Tan W, Zhang Z, Chen X. An automated approach for predicting glioma grade and survival of LGG patients using CNN and radiomics. Front Oncol 2022; 12:969907. [PMID: 36033433 PMCID: PMC9413530 DOI: 10.3389/fonc.2022.969907] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 07/15/2022] [Indexed: 12/24/2022] Open
Abstract
Objectives To develop and validate an efficient and automatically computational approach for stratifying glioma grades and predicting survival of lower-grade glioma (LGG) patients using an integration of state-of-the-art convolutional neural network (CNN) and radiomics. Method This retrospective study reviewed 470 preoperative MR images of glioma from BraTs public dataset (n=269) and Jinling hospital (n=201). A fully automated pipeline incorporating tumor segmentation and grading was developed, which can avoid variability and subjectivity of manual segmentations. First, an integrated approach by fusing CNN features and radiomics features was employed to stratify glioma grades. Then, a deep-radiomics signature based on the integrated approach for predicting survival of LGG patients was developed and subsequently validated in an independent cohort. Results The performance of tumor segmentation achieved a Dice coefficient of 0.81. The intraclass correlation coefficients (ICCs) of the radiomics features between the segmentation network and physicians were all over 0.75. The performance of glioma grading based on integrated approach achieved the area under the curve (AUC) of 0.958, showing the effectiveness of the integrated approach. The multivariable Cox regression results demonstrated that the deep-radiomics signature remained an independent prognostic factor and the integrated nomogram showed significantly better performance than the clinical nomogram in predicting overall survival of LGG patients (C-index: 0.865 vs. 0.796, P=0.005). Conclusion The proposed integrated approach can be noninvasively and efficiently applied in prediction of gliomas grade and survival. Moreover, our fully automated pipeline successfully achieved computerized segmentation instead of manual segmentation, which shows the potential to be a reproducible approach in clinical practice.
Collapse
Affiliation(s)
- Chenan Xu
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, and School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou, China
| | - Yuanyuan Peng
- School of Electronics and Information Engineering and Medical Image Processing, Analysis and Visualization Lab, Soochow University, Suzhou, China
| | - Weifang Zhu
- School of Electronics and Information Engineering and Medical Image Processing, Analysis and Visualization Lab, Soochow University, Suzhou, China
| | - Zhongyue Chen
- School of Electronics and Information Engineering and Medical Image Processing, Analysis and Visualization Lab, Soochow University, Suzhou, China
| | - Jianrui Li
- Department of Diagnostic Radiology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Wenhao Tan
- School of Electronics and Information Engineering and Medical Image Processing, Analysis and Visualization Lab, Soochow University, Suzhou, China
| | - Zhiqiang Zhang
- Department of Diagnostic Radiology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
- State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, China
- *Correspondence: Zhiqiang Zhang, ; Xinjian Chen,
| | - Xinjian Chen
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, and School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou, China
- School of Electronics and Information Engineering and Medical Image Processing, Analysis and Visualization Lab, Soochow University, Suzhou, China
- *Correspondence: Zhiqiang Zhang, ; Xinjian Chen,
| |
Collapse
|
17
|
Xu J, Meng Y, Qiu K, Topatana W, Li S, Wei C, Chen T, Chen M, Ding Z, Niu G. Applications of Artificial Intelligence Based on Medical Imaging in Glioma: Current State and Future Challenges. Front Oncol 2022; 12:892056. [PMID: 35965542 PMCID: PMC9363668 DOI: 10.3389/fonc.2022.892056] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 06/22/2022] [Indexed: 12/24/2022] Open
Abstract
Glioma is one of the most fatal primary brain tumors, and it is well-known for its difficulty in diagnosis and management. Medical imaging techniques such as magnetic resonance imaging (MRI), positron emission tomography (PET), and spectral imaging can efficiently aid physicians in diagnosing, treating, and evaluating patients with gliomas. With the increasing clinical records and digital images, the application of artificial intelligence (AI) based on medical imaging has reduced the burden on physicians treating gliomas even further. This review will classify AI technologies and procedures used in medical imaging analysis. Additionally, we will discuss the applications of AI in glioma, including tumor segmentation and classification, prediction of genetic markers, and prediction of treatment response and prognosis, using MRI, PET, and spectral imaging. Despite the benefits of AI in clinical applications, several issues such as data management, incomprehension, safety, clinical efficacy evaluation, and ethical or legal considerations, remain to be solved. In the future, doctors and researchers should collaborate to solve these issues, with a particular emphasis on interdisciplinary teamwork.
Collapse
Affiliation(s)
- Jiaona Xu
- Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuting Meng
- Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kefan Qiu
- Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Win Topatana
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shijie Li
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chao Wei
- Department of Neurology, Affiliated Ningbo First Hospital, Ningbo, China
| | - Tianwen Chen
- Department of Neurology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Mingyu Chen
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Mingyu Chen, ; Zhongxiang Ding, ; Guozhong Niu,
| | - Zhongxiang Ding
- Department of Radiology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Mingyu Chen, ; Zhongxiang Ding, ; Guozhong Niu,
| | - Guozhong Niu
- Department of Neurology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Mingyu Chen, ; Zhongxiang Ding, ; Guozhong Niu,
| |
Collapse
|
18
|
Jian A, Liu S, Di Ieva A. Artificial Intelligence for Survival Prediction in Brain Tumors on Neuroimaging. Neurosurgery 2022; 91:8-26. [PMID: 35348129 DOI: 10.1227/neu.0000000000001938] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 01/08/2022] [Indexed: 12/30/2022] Open
Abstract
Survival prediction of patients affected by brain tumors provides essential information to guide surgical planning, adjuvant treatment selection, and patient counseling. Current reliance on clinical factors, such as Karnofsky Performance Status Scale, and simplistic radiological characteristics are, however, inadequate for survival prediction in tumors such as glioma that demonstrate molecular and clinical heterogeneity with variable survival outcomes. Advances in the domain of artificial intelligence have afforded powerful tools to capture a large number of hidden high-dimensional imaging features that reflect abundant information about tumor structure and physiology. Here, we provide an overview of current literature that apply computational analysis tools such as radiomics and machine learning methods to the pipeline of image preprocessing, tumor segmentation, feature extraction, and construction of classifiers to establish survival prediction models based on neuroimaging. We also discuss challenges relating to the development and evaluation of such models and explore ethical issues surrounding the future use of machine learning predictions.
Collapse
Affiliation(s)
- Anne Jian
- Computational NeuroSurgery (CNS) Lab, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
- Royal Melbourne Hospital, Melbourne, Australia
| | - Sidong Liu
- Computational NeuroSurgery (CNS) Lab, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
- Centre for Health Informatics, Australian Institute of Health Innovation, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
| | - Antonio Di Ieva
- Computational NeuroSurgery (CNS) Lab, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
| |
Collapse
|
19
|
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: 0] [Impact Index Per Article: 0] [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
|
20
|
Liu ET, Zhou S, Li Y, Zhang S, Ma Z, Guo J, Guo L, Zhang Y, Guo Q, Xu L. Development and validation of an MRI-based nomogram for the preoperative prediction of tumor mutational burden in lower-grade gliomas. Quant Imaging Med Surg 2022; 12:1684-1697. [PMID: 35284257 PMCID: PMC8899970 DOI: 10.21037/qims-21-300] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 08/30/2021] [Indexed: 09/25/2023]
Abstract
BACKGROUND High tumor mutational burden (TMB) is an emerging biomarker of sensitivity to immune checkpoint inhibitors. In this study, we aimed to determine the value of magnetic resonance (MR)-based preoperative nomogram in predicting TMB status in lower-grade glioma (LGG) patients. METHODS Overall survival (OS) data were derived from The Cancer Genome Atlas (TCGA) and then analyzed by using the Kaplan-Meier method and time-dependent receiver operating characteristic (tdROC) analysis. The magnetic resonance imaging (MRI) data of 168 subjects obtained from The Cancer Imaging Archive (TCIA) were retrospectively analyzed. The correlation was explored by univariate and multivariate regression analyses. Finally, we performed tenfold cross validation. TMB values were retrieved from the supplementary information of a previously published article. RESULTS The high TMB subtype was associated with the shortest median OS (high vs. low: 50.9 vs. 95.6 months, P<0.05). The tdROC for the high-TMB tumors was 74% (95% CI: 61-86%) for survival at 12 months, and 71% (95% CI: 60-82%) for survival at 24 months. Multivariate logistic regression analysis confirmed that three risk factors [extranodular growth: odds ratio (OR): 8.367, 95% CI: 3.153-22.199, P<0.01; length-width ratio ≥ median: OR: 1.947, 95% CI: 1.025-3.697, P<0.05; frontal lobe: OR: 0.455, 95% CI: 0.229-0.903, P<0.05] were significant independent predictors of high-TMB tumors. The nomogram showed good calibration and discrimination. This model had an area under the curve (AUC) of 0.736 (95% CI: 0.655-0.817). Decision curve analysis (DCA) demonstrated that the nomogram was clinically useful. The average accuracy of the tenfold cross validation was 71.6% for high-TMB tumors. CONCLUSIONS Our results indicated that a distinct OS disadvantage was associated with the high TMB group. In addition, extranodular growth, nonfrontal lobe tumors and length-width ratio ≥ median can be conveniently used to facilitate the prediction of high-TMB tumors.
Collapse
Affiliation(s)
- En-Tao Liu
- WeiLun PET Center, Department of Nuclear Medicine, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Shuqin Zhou
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine and Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Yingwen Li
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine and Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Siwei Zhang
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine and Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Zelan Ma
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine and Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Junbiao Guo
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine and Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Lei Guo
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine and Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Yue Zhang
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine and Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Quanlai Guo
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine and Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Li Xu
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine and Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| |
Collapse
|
21
|
Tian Y, Komolafe TE, Chen T, Zhou B, Yang X. Prediction of TACE Treatment Response in a Preoperative MRI via Analysis of Integrating Deep Learning and Radiomics Features. J Med Biol Eng 2022. [DOI: 10.1007/s40846-022-00692-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
22
|
Park CJ, Park YW, Ahn SS, Kim D, Kim EH, Kang SG, Chang JH, Kim SH, Lee SK. Quality of Radiomics Research on Brain Metastasis: A Roadmap to Promote Clinical Translation. Korean J Radiol 2022; 23:77-88. [PMID: 34983096 PMCID: PMC8743155 DOI: 10.3348/kjr.2021.0421] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/05/2021] [Accepted: 08/05/2021] [Indexed: 12/14/2022] Open
Abstract
Objective Our study aimed to evaluate the quality of radiomics studies on brain metastases based on the radiomics quality score (RQS), Transparent Reporting of a multivariable prediction model for Individual Prognosis Or Diagnosis (TRIPOD) checklist, and the Image Biomarker Standardization Initiative (IBSI) guidelines. Materials and Methods PubMed MEDLINE, and EMBASE were searched for articles on radiomics for evaluating brain metastases, published until February 2021. Of the 572 articles, 29 relevant original research articles were included and evaluated according to the RQS, TRIPOD checklist, and IBSI guidelines. Results External validation was performed in only three studies (10.3%). The median RQS was 3.0 (range, -6 to 12), with a low basic adherence rate of 50.0%. The adherence rate was low in comparison to the “gold standard” (10.3%), stating the potential clinical utility (10.3%), performing the cut-off analysis (3.4%), reporting calibration statistics (6.9%), and providing open science and data (3.4%). None of the studies involved test-retest or phantom studies, prospective studies, or cost-effectiveness analyses. The overall rate of adherence to the TRIPOD checklist was 60.3% and low for reporting title (3.4%), blind assessment of outcome (0%), description of the handling of missing data (0%), and presentation of the full prediction model (0%). The majority of studies lacked pre-processing steps, with bias-field correction, isovoxel resampling, skull stripping, and gray-level discretization performed in only six (20.7%), nine (31.0%), four (3.8%), and four (13.8%) studies, respectively. Conclusion The overall scientific and reporting quality of radiomics studies on brain metastases published during the study period was insufficient. Radiomics studies should adhere to the RQS, TRIPOD, and IBSI guidelines to facilitate the translation of radiomics into the clinical field.
Collapse
Affiliation(s)
- Chae Jung Park
- Department of Radiology, Research Institute of Radiological Science and Center for Clinical Imaging Data Science, Yongin Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Yae Won Park
- Department of Radiology and Research Institute of Radiological Science and Center for Clinical Imaging Data Science, Yonsei University College of Medicine, Seoul, Korea
| | - Sung Soo Ahn
- Department of Radiology and Research Institute of Radiological Science and Center for Clinical Imaging Data Science, Yonsei University College of Medicine, Seoul, Korea
| | - Dain Kim
- Department of Psychology, Yonsei University, Seoul, Korea
| | - Eui Hyun Kim
- Department of Neurosurgery, Yonsei University College of Medicine, Seoul, Korea
| | - Seok-Gu Kang
- Department of Neurosurgery, Yonsei University College of Medicine, Seoul, Korea
| | - Jong Hee Chang
- Department of Neurosurgery, Yonsei University College of Medicine, Seoul, Korea
| | - Se Hoon Kim
- Department of Pathology, Yonsei University College of Medicine, Seoul, Korea
| | - Seung-Koo Lee
- Department of Radiology and Research Institute of Radiological Science and Center for Clinical Imaging Data Science, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
23
|
Kang CY, Duarte SE, Kim HS, Kim E, Park J, Lee AD, Kim Y, Kim L, Cho S, Oh Y, Gim G, Park I, Lee D, Abazeed M, Velichko YS, Chae YK. OUP accepted manuscript. Oncologist 2022; 27:e471-e483. [PMID: 35348765 PMCID: PMC9177100 DOI: 10.1093/oncolo/oyac036] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 01/14/2022] [Indexed: 11/17/2022] Open
Abstract
The recent, rapid advances in immuno-oncology have revolutionized cancer treatment and spurred further research into tumor biology. Yet, cancer patients respond variably to immunotherapy despite mounting evidence to support its efficacy. Current methods for predicting immunotherapy response are unreliable, as these tests cannot fully account for tumor heterogeneity and microenvironment. An improved method for predicting response to immunotherapy is needed. Recent studies have proposed radiomics—the process of converting medical images into quantitative data (features) that can be processed using machine learning algorithms to identify complex patterns and trends—for predicting response to immunotherapy. Because patients undergo numerous imaging procedures throughout the course of the disease, there exists a wealth of radiological imaging data available for training radiomics models. And because radiomic features reflect cancer biology, such as tumor heterogeneity and microenvironment, these models have enormous potential to predict immunotherapy response more accurately than current methods. Models trained on preexisting biomarkers and/or clinical outcomes have demonstrated potential to improve patient stratification and treatment outcomes. In this review, we discuss current applications of radiomics in oncology, followed by a discussion on recent studies that use radiomics to predict immunotherapy response and toxicity.
Collapse
Affiliation(s)
| | | | - Hye Sung Kim
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Eugene Kim
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | | | - Alice Daeun Lee
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Yeseul Kim
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Leeseul Kim
- Department of Internal Medicine, AMITA Health Saint Francis Hospital, Evanston, IL, USA
| | - Sukjoo Cho
- Department of Pediatrics, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Yoojin Oh
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Gahyun Gim
- Department of Hematology and Oncology, Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Inae Park
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Dongyup Lee
- Department of Physical Medicine and Rehabilitation, Geisinger Health System, Danville, PA, USA
| | - Mohamed Abazeed
- Department of Radiation Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Yury S Velichko
- Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Young Kwang Chae
- Corresponding author: Young Kwang Chae, Department of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
24
|
Tian Y, Komolafe TE, Zheng J, Zhou G, Chen T, Zhou B, Yang X. Assessing PD-L1 Expression Level via Preoperative MRI in HCC Based on Integrating Deep Learning and Radiomics Features. Diagnostics (Basel) 2021; 11:1875. [PMID: 34679573 PMCID: PMC8534850 DOI: 10.3390/diagnostics11101875] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 10/02/2021] [Accepted: 10/04/2021] [Indexed: 12/30/2022] Open
Abstract
To assess if quantitative integrated deep learning and radiomics features can predict the PD-L1 expression level in preoperative MRI of hepatocellular carcinoma (HCC) patients. The data in this study consist of 103 hepatocellular carcinoma patients who received immunotherapy in a single center. These patients were divided into a high PD-L1 expression group (30 patients) and a low PD-L1 expression group (73 patients). Both radiomics and deep learning features were extracted from their MRI sequence of T2-WI, which were merged into an integrative feature space for machine learning for the prediction of PD-L1 expression. The five-fold cross-validation was adopted to validate the performance of the model, while the AUC was used to assess the predictive ability of the model. Based on the five-fold cross-validation, the integrated model achieved the best prediction performance, with an AUC score of 0.897 ± 0.084, followed by the deep learning-based model with an AUC of 0.852 ± 0.043 then the radiomics-based model with AUC of 0.794 ± 0.035. The feature set integrating radiomics and deep learning features is more effective in predicting PD-L1 expression level than only one feature type. The integrated model can achieve fast and accurate prediction of PD-L1 expression status in preoperative MRI of HCC patients.
Collapse
Affiliation(s)
- Yuchi Tian
- Academy of Engineering and Technology, Fudan University, Shanghai 200433, China;
| | | | - Jian Zheng
- Department of Medical Imaging, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China;
| | - Guofeng Zhou
- Department of Radiology, Zhongshan Hospital, Shanghai 200032, China;
| | - Tao Chen
- School of Information Science and Technology, Fudan University, Shanghai 200433, China;
| | - Bo Zhou
- Department of Interventional Radiology, Zhongshan Hospital, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Xiaodong Yang
- Academy of Engineering and Technology, Fudan University, Shanghai 200433, China;
- Department of Medical Imaging, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China;
| |
Collapse
|
25
|
Zhai Y, Song D, Yang F, Wang Y, Jia X, Wei S, Mao W, Xue Y, Wei X. Preoperative Prediction of Meningioma Consistency via Machine Learning-Based Radiomics. Front Oncol 2021; 11:657288. [PMID: 34123812 PMCID: PMC8187861 DOI: 10.3389/fonc.2021.657288] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 04/12/2021] [Indexed: 12/30/2022] Open
Abstract
Objectives The aim of this study was to establish and validate a radiomics nomogram for predicting meningiomas consistency, which could facilitate individualized operation schemes-making. Methods A total of 172 patients was enrolled in the study (train cohort: 120 cases, test cohort: 52 cases). Tumor consistency was classified as soft or firm according to Zada’s consistency grading system. Radiomics features were extracted from multiparametric MRI. Variance selection and LASSO regression were used for feature selection. Then, radiomics models were constructed by five classifiers, and the area under curve (AUC) was used to evaluate the performance of each classifiers. A radiomics nomogram was developed using the best classifier. The performance of this nomogram was assessed by AUC, calibration and discrimination. Results A total of 3840 radiomics features were extracted from each patient, of which 3719 radiomics features were stable features. 28 features were selected to construct the radiomics nomogram. Logistic regression classifier had the highest prediction efficacy. Radiomics nomogram was constructed using logistic regression in the train cohort. The nomogram showed a good sensitivity and specificity with AUCs of 0.861 and 0.960 in train and test cohorts, respectively. Moreover, the calibration graph of the nomogram showed a favorable calibration in both train and test cohorts. Conclusions The presented radiomics nomogram, as a non-invasive prediction tool, could predict meningiomas consistency preoperatively with favorable accuracy, and facilitated the determination of individualized operation schemes.
Collapse
Affiliation(s)
- Yixuan Zhai
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Dixiang Song
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Fengdong Yang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yiming Wang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xin Jia
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shuxin Wei
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wenbin Mao
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yake Xue
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xinting Wei
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
26
|
Li Q, Dong F, Jiang B, Zhang M. Exploring MRI Characteristics of Brain Diffuse Midline Gliomas With the H3 K27M Mutation Using Radiomics. Front Oncol 2021; 11:646267. [PMID: 34109112 PMCID: PMC8182051 DOI: 10.3389/fonc.2021.646267] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Accepted: 04/26/2021] [Indexed: 01/01/2023] Open
Abstract
Objectives To explore the magnetic resonance imaging (MRI) characteristics of brain diffuse midline gliomas with the H3 K27M mutation (DMG-M) using radiomics. Materials and Methods Thirty patients with diffuse midline gliomas, including 16 with the H3 K27M mutant and 14 with wild type tumors, were retrospectively included in this study. A total of 272 radiomic features were initially extracted from MR images of each tumor. Principal component analysis, univariate analysis, and three other feature selection methods, including variance thresholding, recursive feature elimination, and the elastic net, were used to analyze the radiomic features. Based on the results, related visually accessible features of the tumors were further evaluated. Results Patients with DMG-M were younger than those with diffuse midline gliomas with H3 K27M wild (DMG-W) (median, 25.5 and 48 years old, respectively; p=0.005). Principal component analysis showed that there were obvious overlaps in the first two principal components for both DMG-M and DMG-W tumors. The feature selection results showed that few features from T2-weighted images (T2WI) were useful for differentiating DMG-M and DMG-W tumors. Thereafter, four visually accessible features related to T2WI were further extracted and analyzed. Among these features, only cystic formation showed a significant difference between the two types of tumors (OR=7.800, 95% CI 1.476-41.214, p=0.024). Conclusions DMGs with and without the H3 K27M mutation shared similar MRI characteristics. T2W sequences may be valuable, and cystic formation a useful MRI biomarker, for diagnosing brain DMG-M.
Collapse
Affiliation(s)
- Qian Li
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Fei Dong
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Biao Jiang
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Minming Zhang
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
27
|
Habib A, Jovanovich N, Hoppe M, Ak M, Mamindla P, R. Colen R, Zinn PO. MRI-Based Radiomics and Radiogenomics in the Management of Low-Grade Gliomas: Evaluating the Evidence for a Paradigm Shift. J Clin Med 2021; 10:1411. [PMID: 33915813 PMCID: PMC8036428 DOI: 10.3390/jcm10071411] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/23/2021] [Accepted: 03/25/2021] [Indexed: 12/29/2022] Open
Abstract
Low-grade gliomas (LGGs) are tumors that affect mostly adults. These neoplasms are comprised mainly of oligodendrogliomas and diffuse astrocytomas. LGGs remain vexing to current management and therapeutic modalities although they exhibit more favorable survival rates compared with high-grade gliomas (HGGs). The specific genetic subtypes that these tumors exhibit result in variable clinical courses and the need to involve multidisciplinary teams of neurologists, epileptologists, neurooncologists and neurosurgeons. Currently, the diagnosis of an LGG pivots mainly around the preliminary radiological findings and the subsequent definitive surgical diagnosis (via surgical sampling). The introduction of radiomics as a high throughput quantitative imaging technique that allows for improved diagnostic, prognostic and predictive indices has created more interest for such techniques in cancer research and especially in neurooncology (MRI-based classification of LGGs, predicting Isocitrate dehydrogenase (IDH) and Telomerase reverse transcriptase (TERT) promoter mutations and predicting LGG associated seizures). Radiogenomics refers to the linkage of imaging findings with the tumor/tissue genomics. Numerous applications of radiomics and radiogenomics have been described in the clinical context and management of LGGs. In this review, we describe the recently published studies discussing the potential application of radiomics and radiogenomics in LGGs. We also highlight the potential pitfalls of the above-mentioned high throughput computerized techniques and, most excitingly, explore the use of machine learning artificial intelligence technologies as standalone and adjunct imaging tools en route to enhance a personalized MRI-based tumor diagnosis and management plan design.
Collapse
Affiliation(s)
- Ahmed Habib
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15232, USA;
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA 15232, USA; (N.J.); (M.H.); (M.A.); (P.M.); (R.R.C.)
| | - Nicolina Jovanovich
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA 15232, USA; (N.J.); (M.H.); (M.A.); (P.M.); (R.R.C.)
| | - Meagan Hoppe
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA 15232, USA; (N.J.); (M.H.); (M.A.); (P.M.); (R.R.C.)
| | - Murat Ak
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA 15232, USA; (N.J.); (M.H.); (M.A.); (P.M.); (R.R.C.)
- Department of Diagnostic Radiology, University of Pittsburgh Medical Center, Pittsburgh, PA 15232, USA
| | - Priyadarshini Mamindla
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA 15232, USA; (N.J.); (M.H.); (M.A.); (P.M.); (R.R.C.)
| | - Rivka R. Colen
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA 15232, USA; (N.J.); (M.H.); (M.A.); (P.M.); (R.R.C.)
- Department of Diagnostic Radiology, University of Pittsburgh Medical Center, Pittsburgh, PA 15232, USA
| | - Pascal O. Zinn
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15232, USA;
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA 15232, USA; (N.J.); (M.H.); (M.A.); (P.M.); (R.R.C.)
| |
Collapse
|
28
|
Radiomics for prediction of survival in lower-grade gliomas-it's time to move beyond the crystal ball. Eur Radiol 2020; 31:1783-1784. [PMID: 33341906 PMCID: PMC7979609 DOI: 10.1007/s00330-020-07603-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 12/04/2020] [Indexed: 11/26/2022]
Abstract
• Radiomics might help predict survival of patients with lower-grade gliomas. • Several different models using different radiomics features have been proposed with only little overlap in included features. • Prospective trials and validation studies are needed to establish which models offer clinical benefit and which do not.
Collapse
|