1
|
Fares J, Wan Y, Mayrand R, Li Y, Mair R, Price SJ. Decoding Glioblastoma Heterogeneity: Neuroimaging Meets Machine Learning. Neurosurgery 2025; 96:1181-1192. [PMID: 39570018 PMCID: PMC12052239 DOI: 10.1227/neu.0000000000003260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 09/18/2024] [Indexed: 11/22/2024] Open
Abstract
Recent advancements in neuroimaging and machine learning have significantly improved our ability to diagnose and categorize isocitrate dehydrogenase (IDH)-wildtype glioblastoma, a disease characterized by notable tumoral heterogeneity, which is crucial for effective treatment. Neuroimaging techniques, such as diffusion tensor imaging and magnetic resonance radiomics, provide noninvasive insights into tumor infiltration patterns and metabolic profiles, aiding in accurate diagnosis and prognostication. Machine learning algorithms further enhance glioblastoma characterization by identifying distinct imaging patterns and features, facilitating precise diagnoses and treatment planning. Integration of these technologies allows for the development of image-based biomarkers, potentially reducing the need for invasive biopsy procedures and enabling personalized therapy targeting specific pro-tumoral signaling pathways and resistance mechanisms. Although significant progress has been made, ongoing innovation is essential to address remaining challenges and further improve these methodologies. Future directions should focus on refining machine learning models, integrating emerging imaging techniques, and elucidating the complex interplay between imaging features and underlying molecular processes. This review highlights the pivotal role of neuroimaging and machine learning in glioblastoma research, offering invaluable noninvasive tools for diagnosis, prognosis prediction, and treatment planning, ultimately improving patient outcomes. These advances in the field promise to usher in a new era in the understanding and classification of IDH-wildtype glioblastoma.
Collapse
Affiliation(s)
- Jawad Fares
- Department of Clinical Neurosciences, Academic Neurosurgery Division, University of Cambridge, Cambridge, UK
- Cambridge Brain Tumour Imaging Laboratory, Department of Clinical Neurosciences, Academic Neurosurgery Division, University of Cambridge, Cambridge, UK
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Yizhou Wan
- Department of Clinical Neurosciences, Academic Neurosurgery Division, University of Cambridge, Cambridge, UK
- Cambridge Brain Tumour Imaging Laboratory, Department of Clinical Neurosciences, Academic Neurosurgery Division, University of Cambridge, Cambridge, UK
| | - Roxanne Mayrand
- Department of Clinical Neurosciences, Academic Neurosurgery Division, University of Cambridge, Cambridge, UK
- Cambridge Brain Tumour Imaging Laboratory, Department of Clinical Neurosciences, Academic Neurosurgery Division, University of Cambridge, Cambridge, UK
| | - Yonghao Li
- Department of Clinical Neurosciences, Academic Neurosurgery Division, University of Cambridge, Cambridge, UK
- Cambridge Brain Tumour Imaging Laboratory, Department of Clinical Neurosciences, Academic Neurosurgery Division, University of Cambridge, Cambridge, UK
| | - Richard Mair
- Department of Clinical Neurosciences, Academic Neurosurgery Division, University of Cambridge, Cambridge, UK
| | - Stephen J. Price
- Department of Clinical Neurosciences, Academic Neurosurgery Division, University of Cambridge, Cambridge, UK
- Cambridge Brain Tumour Imaging Laboratory, Department of Clinical Neurosciences, Academic Neurosurgery Division, University of Cambridge, Cambridge, UK
| |
Collapse
|
2
|
Liang F, Cao Y, Zhao T, Xu Q, Zhu H. Semisupervised adaptive learning models for IDH1 mutation status prediction. PLoS One 2025; 20:e0321404. [PMID: 40323991 PMCID: PMC12052117 DOI: 10.1371/journal.pone.0321404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 03/05/2025] [Indexed: 05/07/2025] Open
Abstract
The mutation status of isocitrate dehydrogenase1 (IDH1) in glioma is critical information for the diagnosis, treatment, and prognosis. Accurately determining such information from MRI data has emerged as a significant research challenge in recent years. Existing techniques for this problem often suffer from various limitations, such as the data waste and instability issues. To address such issues, we present a semisupervised adaptive deep learning model based on radiomics and rough sets for predicting the mutation status of IDH1 from MRI data. Firstly, our model uses a rough set algorithm to remove the redundant medical image features extracted by radiomics, while adding pseudo-labels for non-labeled data via statistical. T-tests to mitigate the common issue of insufficient datasets in medical imaging analysis. Then, it applies a Sand Cat Swarm Optimization (SCSO) algorithm to optimize the weight of pseudo-label data. Finally, our model adopts U-Net and CRNN to construct UCNet, a semisupervised classification model for classifying IDH1 mutation status. To validate our models, we use a preoperative MRI dataset with 316 glioma patients to evaluate the performance. Our study suggests that the prediction accuracy of glioma IDH1 mutation status reaches 95.63%. Our experimental results suggest that the study can effectively improve the utilization of glioma imaging data and the accuracy of intelligent diagnosis of glioma IDH1 mutation status.
Collapse
Affiliation(s)
- Fengning Liang
- School of Medical Information and Engineering, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yaru Cao
- School of Medical Information and Engineering, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Teng Zhao
- School of Medical Information and Engineering, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Qian Xu
- Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Hong Zhu
- School of Medical Information and Engineering, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Computer Science and Engineering, State University of New York at Buffalo, Buffalo, New York, United States of America
| |
Collapse
|
3
|
Huang Z, Qiu Z, Chen S, Zhang Y, Wang K, Zeng Q, Huang Y, Zhang Y, Bu J. Optimizing Deep Learning Models for Luminal and Nonluminal Breast Cancer Classification Using Multidimensional ROI in DCE-MRI-A Multicenter Study. Cancer Med 2025; 14:e70931. [PMID: 40347080 PMCID: PMC12065080 DOI: 10.1002/cam4.70931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 04/14/2025] [Accepted: 04/25/2025] [Indexed: 05/12/2025] Open
Abstract
OBJECTIVES Previous deep learning studies have not explored the synergistic effects of ROI dimensions (2D/2.5D/3D), peritumoral expansion levels (0-8 mm), and segmentation scenarios (ROI only vs. ROI original). Our study aims to evaluate the performance of multidimensional deep transfer learning models in distinguishing molecular subtypes of breast cancer (luminal vs. nonluminal) using DCE-MRI. Under two segmentation scenarios, we systematically compare the effects of ROI dimensions and peritumoral expansion levels to optimize multidimensional deep learning models via transfer learning for distinguishing luminal from nonluminal breast cancers in DCE-MRI-based analysis. MATERIALS AND METHODS From October 2020 to October 2023, data from 426 patients with primary invasive breast cancer were retrospectively collected. Patients were divided into three cohorts: (1) training cohort, n = 108, from SYSU Hospital (Zhuhai, China); (2) validation cohort 1, n = 165, from HZ Hospital (Huizhou, China); and (3) validation cohort 2, n = 153, from LY Hospital (Linyi, China). ROIs were delineated, and expansions of 2, 4, 6, and 8 mm beyond the lesion boundary were performed. We assessed the performance of various deep transfer learning models, considering precise segmentation (ROI only and ROI original) and varying peritumoral regions, using ROC curves and decision curve analysis. RESULTS The 2.5D1-based deep learning model (ROI original, 4 mm expansion) demonstrated optimal performance, achieving an AUC of 0.808 (95% CI 0.715-0.901) in the training cohort, 0.766 (95% CI 0.682-0.850) in validation cohort 1, and 0.799 (95% CI 0.725-0.874) in validation cohort 2. CONCLUSION The study highlights that the 2.5D1-based deep learning model utilizing the three principal slices of the minimum bounding box (ROI original) with a 4 mm peritumoral region is effective in distinguishing between luminal and nonluminal breast cancer tumors, serving as a potential diagnostic tool.
Collapse
Affiliation(s)
- Zhenfeng Huang
- Department of Thyroid & Breast SurgeryThe Fifth Affiliated Hospital, Sun Yat‐sen UniversityZhuhaiChina
- Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated HospitalSun Yat‐sen UniversityZhuhaiChina
| | - Zhikun Qiu
- Department of Breast SurgeryHuizhou Central People's HospitalHuizhouChina
| | | | - Yideng Zhang
- Department of Thyroid & Breast SurgeryThe Fifth Affiliated Hospital, Sun Yat‐sen UniversityZhuhaiChina
| | - Kunyi Wang
- Department of Thyroid & Breast SurgeryThe Fifth Affiliated Hospital, Sun Yat‐sen UniversityZhuhaiChina
| | - Qingan Zeng
- Department of Thyroid & Breast SurgeryThe Fifth Affiliated Hospital, Sun Yat‐sen UniversityZhuhaiChina
| | - Yukang Huang
- Department of Breast SurgeryHuizhou Central People's HospitalHuizhouChina
| | | | - Juyuan Bu
- Department of Gastrointestinal Surgery, the Fifth Affiliated HospitalSun Yat‐Sen UniversityZhuhaiChina
| |
Collapse
|
4
|
Jian J, Xu L, Gong C, Ding S, Gong X, Yuan X, Zheng W, Wang X, Zhang Y. One class classification-empowered radiomics for noninvasively accurate prediction of glioma isocitrate dehydrogenase mutation using multiparametric magnetic resonance imaging. Clin Radiol 2025; 84:106866. [PMID: 40157017 DOI: 10.1016/j.crad.2025.106866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 02/20/2025] [Accepted: 03/02/2025] [Indexed: 04/01/2025]
Abstract
BACKGROUND Noninvasive detection of isocitrate dehydrogenase (IDH) mutations is crucial for preoperative decision-making in patients with glioma. While radiomics has been applied, data imbalance-specifically between IDH wild-type and mutated genes-remains underexplored. We developed a one-class classification-empowered radiomics (OCCR) model, trained exclusively on IDH wild-type patients, to distinguish them from IDH mutation cases. MATERIALS AND METHODS This study included 495 patients from the UCSF Preoperative Diffuse Glioma MRI dataset. T1, T1ce, and FLAIR sequences were registered to T2 and resampled to a 1-mm isotropic resolution. The coregistered data were skull-stripped, and the tumor region was segmented using an ensemble model, followed by manual refinement. We extracted 386 radiomics features from the four MRI sequences and input them into an auto-encoder with 7 hidden layers for reconstruction. The OCCR model was trained on wild-type IDH patients, using the mean square error between the original and reconstructed features as guidance. During validation, reconstruction error was used to differentiate IDH mutations from the wild type. RESULTS The hold-out validation demonstrated that OCCR performance improved as the number of training samples increased, achieving a peak area under the receiver operating characteristic curve of 0.8018. Visualization of reconstruction errors highlighted first-order and gray-level co-occurrence matrix features in the T1ce sequence. CONCLUSIONS This study demonstrates the feasibility of integrating one-class classification into radiomics for the determination of preoperative IDH mutation status in patients with glioma using multiparametric MRI. This versatile model holds potential for other diseases with substantial data imbalance.
Collapse
Affiliation(s)
- J Jian
- Department of Radiation Oncology, Jiangxi Cancer Hospital&Institute, Nanchang, Jiangxi, PR China; Jiangxi Clinical Research Center for Cancer, Nanchang, Jiangxi, PR China; The Second Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, PR China; NHC Key Laboratory of Personalized Diagnosis and Treatment of Nasopharyngeal Carcinoma, Nanchang, Jiangxi, PR China
| | - L Xu
- Department of Radiation Oncology, Jiangxi Cancer Hospital&Institute, Nanchang, Jiangxi, PR China
| | - C Gong
- Department of Radiation Oncology, Jiangxi Cancer Hospital&Institute, Nanchang, Jiangxi, PR China
| | - S Ding
- Department of Radiation Oncology, Jiangxi Cancer Hospital&Institute, Nanchang, Jiangxi, PR China
| | - X Gong
- Department of Radiation Oncology, Jiangxi Cancer Hospital&Institute, Nanchang, Jiangxi, PR China
| | - X Yuan
- Department of Radiation Oncology, Jiangxi Cancer Hospital&Institute, Nanchang, Jiangxi, PR China
| | - W Zheng
- Department of Radiation Oncology, Jiangxi Cancer Hospital&Institute, Nanchang, Jiangxi, PR China
| | - X Wang
- Department of Radiation Oncology, Jiangxi Cancer Hospital&Institute, Nanchang, Jiangxi, PR China
| | - Y Zhang
- Department of Radiation Oncology, Jiangxi Cancer Hospital&Institute, Nanchang, Jiangxi, PR China; Jiangxi Clinical Research Center for Cancer, Nanchang, Jiangxi, PR China; The Second Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, PR China; NHC Key Laboratory of Personalized Diagnosis and Treatment of Nasopharyngeal Carcinoma, Nanchang, Jiangxi, PR China; Jiangxi Key Laboratory of Oncology (2024SSY06041), Nanchang, Jiangxi 330029, PR China.
| |
Collapse
|
5
|
Xu L, Li M, Dong X, Wang Z, Tong Y, Feng T, Xu S, Shang H, Zhao B, Lin J, Cao Z, Zheng Y. The value of deep learning and radiomics models in predicting preoperative serosal invasion in gastric cancer: a dual-center study. Abdom Radiol (NY) 2025:10.1007/s00261-025-04949-1. [PMID: 40285792 DOI: 10.1007/s00261-025-04949-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 04/06/2025] [Accepted: 04/09/2025] [Indexed: 04/29/2025]
Abstract
PURPOSE To establish and validate a model based on deep learning (DL), integrating radiomic features with relevant clinical features to generate nomogram, for predicting preoperative serosal invasion in gastric cancer (GC). METHODS This retrospective study included 335 patients from dual centers. T staging (T1-3 or T4) was used to assess serosal invasion. Radiomic features were extracted from primary GC lesions in the venous phase CT, and DL features from 8 transfer learning models were combined to create the Hand-crafted Radiomics and Deep Learning Radiomics (HCR-DLR) model. The Clinical (CL) model was built using clinical features, and both were combined into the Clinical and Radiomics Combined (CRC) model. In total, 15 predictive models were developed using 5 machine learning algorithms. The best-performing models were visualized as nomograms. RESULTS The total of 14 radiomic features, 13 DL features, and 2 clinical features were considered valuable through dimensionality reduction and selection. Among the constructed models: CRC model (AUC, training cohort: 0.9212; internal test cohort: 0.8743; external test cohort: 0.8853) than HCR-DLR model (AUC, training cohort: 0.8607; internal test cohort: 0.8543; external test cohort: 0.8824) and CL model (AUC, training cohort: 0.7632; internal test cohort: 0.7219; external test cohort: 0.7294) showed better performance. A nomogram based on the logistic CL model was drawn to facilitate the usage and showed its excellent predictive performance. CONCLUSION The predictive performance of the CRC Model, which integrates clinical features, radiomic features, and DL features, exhibits robust predictive capability and can serve as a simple, non-invasive, and practical tool for predicting the serosal invasion status of GC.
Collapse
Affiliation(s)
- Lihang Xu
- Radiology Department, Affiliated Hospital of Chengde Medical College, Chengde, China
| | - Mingyu Li
- Radiology Department, Affiliated Hospital of Chengde Medical College, Chengde, China
| | - Xianling Dong
- Hebei International Research Center for Medical-Engineering, Chengde Medical University, Chengde, China
| | - Zhongxiao Wang
- Hebei International Research Center for Medical-Engineering, Chengde Medical University, Chengde, China
| | - Ying Tong
- Radiology Department, Affiliated Hospital of Chengde Medical College, Chengde, China
| | - Tao Feng
- Radiology Department, Affiliated Hospital of Chengde Medical College, Chengde, China
| | - Shuangyan Xu
- Radiology Department, Affiliated Hospital of Chengde Medical College, Chengde, China
| | - Hui Shang
- Radiology Department, Affiliated Hospital of Chengde Medical College, Chengde, China
| | - Bin Zhao
- Radiology Department, Affiliated Hospital of Chengde Medical College, Chengde, China
| | - Jianpeng Lin
- Hebei International Research Center for Medical-Engineering, Chengde Medical University, Chengde, China
| | - Zhendong Cao
- Radiology Department, Affiliated Hospital of Chengde Medical College, Chengde, China.
| | - Yi Zheng
- Radiology department, Chengde Central Hospital, Chengde, China.
| |
Collapse
|
6
|
Gao X, Zhang M, Li J, Zhao S, Zhuo Z, Qu L, Weng J, Chai L, Duan Y, Ye C, Liu Y. Anatomy-guided slice-description interaction for multimodal brain disease diagnosis based on 3D image and radiological report. Comput Med Imaging Graph 2025; 123:102556. [PMID: 40300226 DOI: 10.1016/j.compmedimag.2025.102556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 03/19/2025] [Accepted: 04/13/2025] [Indexed: 05/01/2025]
Abstract
Accurate brain disease diagnosis based on radiological images is desired in clinical practice as it can facilitate early intervention and reduce the risk of damage. However, existing unimodal image-based models struggle to process high-dimensional 3D brain imaging data effectively. Multimodal disease diagnosis approaches based on medical images and corresponding radiological reports achieved promising progress with the development of vision-language models. However, most multimodal methods handle 2D images and cannot be directly applied to brain disease diagnosis that uses 3D images. Therefore, in this work we develop a multimodal brain disease diagnosis model that takes 3D brain images and their radiological reports as input. Motivated by the fact that radiologists scroll through image slices and write important descriptions into the report accordingly, we propose a slice-description cross-modality interaction mechanism to realize fine-grained multimodal data interaction. Moreover, since previous medical research has demonstrated potential correlation between anatomical location of anomalies and diagnosis results, we further explore the use of brain anatomical prior knowledge to improve the multimodal interaction. Based on the report description, the prior knowledge filters the image information by suppressing irrelevant regions and enhancing relevant slices. Our method was validated with two brain disease diagnosis tasks. The results indicate that our model outperforms competing unimodal and multimodal methods for brain disease diagnosis. In particular, it has yielded an average accuracy improvement of 15.87% and 7.39% compared with the image-based and multimodal competing methods, respectively.
Collapse
Affiliation(s)
- Xin Gao
- School of Computer Science, Beijing Institute of Technology, 5 South Zhongguancun Street, Haidian District, Beijing, 100081, China
| | - Meihui Zhang
- School of Computer Science, Beijing Institute of Technology, 5 South Zhongguancun Street, Haidian District, Beijing, 100081, China.
| | - Junjie Li
- Department of Radiology, Beijing Tiantan Hospital, 6 Tiantan Xili, Dongcheng District, Beijing, 100070, China
| | - Shanbo Zhao
- School of Computer Science, Beijing Institute of Technology, 5 South Zhongguancun Street, Haidian District, Beijing, 100081, China
| | - Zhizheng Zhuo
- Department of Radiology, Beijing Tiantan Hospital, 6 Tiantan Xili, Dongcheng District, Beijing, 100070, China
| | - Liying Qu
- Department of Radiology, Beijing Tiantan Hospital, 6 Tiantan Xili, Dongcheng District, Beijing, 100070, China
| | - Jinyuan Weng
- Philips Healthcare, Philips (China) Investment Co. Ltd., Building 718, Lingshi Road, Jingan District, Shanghai, 200072, China
| | - Li Chai
- Department of Radiology, Beijing Tiantan Hospital, 6 Tiantan Xili, Dongcheng District, Beijing, 100070, China
| | - Yunyun Duan
- Department of Radiology, Beijing Tiantan Hospital, 6 Tiantan Xili, Dongcheng District, Beijing, 100070, China
| | - Chuyang Ye
- School of Integrated Circuits and Electronics, Beijing Institute of Technology, 5 Zhongguancun South Street, Haidian District, Beijing, 100081, China.
| | - Yaou Liu
- Department of Radiology, Beijing Tiantan Hospital, 6 Tiantan Xili, Dongcheng District, Beijing, 100070, China.
| |
Collapse
|
7
|
Cui Y, Feng J. One solid step to general neuroradiology AI. Eur Radiol 2025; 35:1933-1934. [PMID: 39179801 DOI: 10.1007/s00330-024-11020-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 07/01/2024] [Accepted: 07/19/2024] [Indexed: 08/26/2024]
Affiliation(s)
- Yuanyuan Cui
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, China.
| | - Jie Feng
- Department of Radiology, Corps Hospital of Shanxi province of Chinese People's Armed Police Force, Taiyuan, China.
| |
Collapse
|
8
|
Li D, Hu W, Ma L, Yang W, Liu Y, Zou J, Ge X, Han Y, Gan T, Cheng D, Ai K, Liu G, Zhang J. Deep learning radiomics nomograms predict Isocitrate dehydrogenase (IDH) genotypes in brain glioma: A multicenter study. Magn Reson Imaging 2025; 117:110314. [PMID: 39708927 DOI: 10.1016/j.mri.2024.110314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 12/16/2024] [Accepted: 12/16/2024] [Indexed: 12/23/2024]
Abstract
PURPOSE To explore the feasibility of Deep learning radiomics nomograms (DLRN) in predicting IDH genotype. METHODS A total of 402 glioma patients from two independent centers were retrospectively included, and the data from center I was randomly divided into a training cohort (n = 239) and an internal validation cohort (n = 103) on a 7:3 basis. Center II served as an independent external validation cohort (n = 60). We developed a DLRN for IDH classification of gliomas based on T2 images. This hybrid model integrates deep learning features, radiomics features, and clinical features most relevant to IDH genotypes and finally classifies them using multivariate logistic regression analysis. We used the area under the curve (AUC) of the receiver operating characteristic (ROC) to evaluate the performance of the model and applied the DLRN score to the survival analysis of some of the follow-up glioma patients. RESULTS The proposed model had an area under the curve (AUC) of 0.98 in an externally validated cohort, and DLRN scores were significantly associated with the overall survival of glioma patients. CONCLUSIONS Deep learning radiomics nomograms performed well in non-invasively predicting IDH mutation status in gliomas, assisting stratified management and targeted therapy for glioma patients.
Collapse
Affiliation(s)
- Darui Li
- Department of Nuclear Magnetic Resonance, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; Gansu Province Clinical Research Center for Functional and Molecular Imaging, Lanzhou 730030, China
| | - Wanjun Hu
- Department of Nuclear Magnetic Resonance, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; Gansu Province Clinical Research Center for Functional and Molecular Imaging, Lanzhou 730030, China
| | - Laiyang Ma
- Department of Nuclear Magnetic Resonance, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; Gansu Province Clinical Research Center for Functional and Molecular Imaging, Lanzhou 730030, China
| | - Wenxia Yang
- Department of Nuclear Magnetic Resonance, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; Gansu Province Clinical Research Center for Functional and Molecular Imaging, Lanzhou 730030, China
| | - Yang Liu
- Department of Nuclear Magnetic Resonance, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; Gansu Province Clinical Research Center for Functional and Molecular Imaging, Lanzhou 730030, China
| | - Jie Zou
- Department of Nuclear Magnetic Resonance, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; Gansu Province Clinical Research Center for Functional and Molecular Imaging, Lanzhou 730030, China
| | - Xin Ge
- Department of Nuclear Magnetic Resonance, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; Gansu Province Clinical Research Center for Functional and Molecular Imaging, Lanzhou 730030, China
| | - Yuping Han
- Department of Nuclear Magnetic Resonance, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; Gansu Province Clinical Research Center for Functional and Molecular Imaging, Lanzhou 730030, China
| | - Tiejun Gan
- Department of Nuclear Magnetic Resonance, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; Gansu Province Clinical Research Center for Functional and Molecular Imaging, Lanzhou 730030, China
| | - Dan Cheng
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Kai Ai
- Philips Healthcare, Xi'an, China
| | - Guangyao Liu
- Department of Nuclear Magnetic Resonance, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; Gansu Province Clinical Research Center for Functional and Molecular Imaging, Lanzhou 730030, China
| | - Jing Zhang
- Department of Nuclear Magnetic Resonance, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; Gansu Province Clinical Research Center for Functional and Molecular Imaging, Lanzhou 730030, China.
| |
Collapse
|
9
|
Wang Y, Hu Z, Wang H. The clinical implications and interpretability of computational medical imaging (radiomics) in brain tumors. Insights Imaging 2025; 16:77. [PMID: 40159380 PMCID: PMC11955438 DOI: 10.1186/s13244-025-01950-6] [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: 10/17/2024] [Accepted: 03/06/2025] [Indexed: 04/02/2025] Open
Abstract
Radiomics has widespread applications in the field of brain tumor research. However, radiomic analyses often function as a 'black box' due to their use of complex algorithms, which hinders the translation of brain tumor radiomics into clinical applications. In this review, we will elaborate extensively on the application of radiomics in brain tumors. Additionally, we will address the interpretability of handcrafted-feature radiomics and deep learning-based radiomics by integrating biological domain knowledge of brain tumors with interpretability methods. Furthermore, we will discuss the current challenges and prospects concerning the interpretability of brain tumor radiomics. Enhancing the interpretability of radiomics may make it more understandable for physicians, ultimately facilitating its translation into clinical practice. CRITICAL RELEVANCE STATEMENT: The interpretability of brain tumor radiomics empowers neuro-oncologists to make well-informed decisions from radiomic models. KEY POINTS: Radiomics makes a significant impact on the management of brain tumors in several key clinical areas. Transparent models, habitat analysis, and feature attribute explanations can enhance the interpretability of traditional handcrafted-feature radiomics in brain tumors. Various interpretability methods have been applied to explain deep learning-based models; however, there is a lack of biological mechanisms underlying these models.
Collapse
Affiliation(s)
- Yixin Wang
- Department of Brain Oncology, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, P. R. China
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, P. R. China
| | - Zongtao Hu
- Department of Brain Oncology, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, P. R. China
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, P. R. China
| | - Hongzhi Wang
- Department of Brain Oncology, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, P. R. China.
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, P. R. China.
| |
Collapse
|
10
|
Yang X, Niu W, Wu K, Yang G, Zhang H. MRI-based habitat imaging predicts high-risk molecular subtypes and early risk assessment of lower-grade gliomas. Cancer Imaging 2025; 25:43. [PMID: 40155946 PMCID: PMC11951782 DOI: 10.1186/s40644-025-00838-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 02/07/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND In lower-grade gliomas (LrGGs, histological grades 2-3), there exist a minority of high-risk molecular subtypes with malignant transformation potential, associated with unfavorable clinical outcomes and shorter survival prognosis. Identifying high-risk molecular subtypes early in LrGGs and conducting preoperative prognostic evaluations are crucial for precise clinical diagnosis and treatment. MATERIALS AND METHODS We retrospectively collected data from 345 patients with LrGGs and comprehensively screened key high-risk molecular markers. Based on preoperative MRI sequences (CE-T1WI/T2-FLAIR), we employed seven classifiers to construct models based on habitat, radiomics, and combined. Eventually, we identified Extra Trees based on habitat features as the optimal predictive model for identifying high-risk molecular subtypes of LrGGs. Moreover, we developed a prognostic prediction model based on radiomics score (Radscore) to assess the survival outlook of patients with LrGGs. We utilized Kaplan-Meier (KM) survival analysis alongside the log-rank test to discern variations in survival probabilities among high-risk and low-risk cohorts. The concordance index was employed to gauge the efficacy of habitat, clinical, and amalgamated prognosis models. Calibration curves were utilized to appraise the congruence between the anticipated survival probability and the actual survival probability projected by the models. RESULTS The habitat model for predicting high-risk molecular subtypes of LrGGs, achieved AUCs of 0.802, 0.771, and 0.768 in the training set, internal test set, and external test set, respectively. Comparison among habitat, clinical, combined prognostic models revealed that the combined prognostic model exhibited the highest performance (C-index = 0.781 in the training set, C-index = 0.778 in the internal test set, C-index = 0.743 in the external test set), followed by the habitat prognostic model (C-index = 0.749 in the training set, C-index = 0.716 in the internal test set, C-index = 0.707 in the external test set), while the clinical prognostic model performed the worst (C-index = 0.717 in the training set, C-index = 0.687 in the internal test set, C-index = 0.649 in the external test set). Furthermore, the calibration curves of the combined model exhibited satisfactory alignment when forecasting the 1-year, 2-year, and 3-year survival probabilities of patients with LrGGs. CONCLUSION The MRI-based habitat model simultaneously achieves the objectives of non-invasive prediction of high-risk molecular subtypes of LrGGs and assessment of survival prognosis. This has incremental value for early non-invasive warning of malignant transformation in LrGGs and risk-stratified management.
Collapse
Affiliation(s)
- Xiangli Yang
- Department of Radiology, First Hospital of Shanxi Medical University, Taiyuan, 030001, China
- Shanxi Bethune Hospital, Third Hospital of Shanxi Medical University, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
- College of Medical Imaging, Shanxi Medical University, Taiyuan, 030001, China
| | - Wenju Niu
- College of Medical Imaging, Shanxi Medical University, Taiyuan, 030001, China
| | - Kai Wu
- Department of Information Management, First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Guoqiang Yang
- Department of Radiology, First Hospital of Shanxi Medical University, Taiyuan, 030001, China.
- College of Medical Imaging, Shanxi Medical University, Taiyuan, 030001, China.
- Shanxi Key Laboratory of Intelligent Imaging and Nanomedicine, First Hospital of Shanxi Medical University, Taiyuan, 030001, China.
| | - Hui Zhang
- Department of Radiology, First Hospital of Shanxi Medical University, Taiyuan, 030001, China.
- College of Medical Imaging, Shanxi Medical University, Taiyuan, 030001, China.
- Shanxi Key Laboratory of Intelligent Imaging and Nanomedicine, First Hospital of Shanxi Medical University, Taiyuan, 030001, China.
- Intelligent Imaging Big Data and Functional Nano-imaging Engineering Research Center of Shanxi Province, First Hospital of Shanxi Medical University, Taiyuan, 030001, China.
| |
Collapse
|
11
|
Niu W, Yan J, Hao M, Zhang Y, Li T, Liu C, Li Q, Liu Z, Su Y, Peng B, Tan Y, Wang X, Wang L, Zhang H, Yang G. MRI transformer deep learning and radiomics for predicting IDH wild type TERT promoter mutant gliomas. NPJ Precis Oncol 2025; 9:89. [PMID: 40148588 PMCID: PMC11950645 DOI: 10.1038/s41698-025-00884-y] [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: 12/01/2024] [Accepted: 03/17/2025] [Indexed: 03/29/2025] Open
Abstract
This study aims to predict IDH wt with TERTp-mut gliomas using multiparametric MRI sequences through a novel fusion model, while matching model classification metrics with patient risk stratification aids in crafting personalized diagnostic and prognosis evaluations.Preoperative T1CE and T2FLAIR sequences from 1185 glioma patients were analyzed. A MultiChannel_2.5D_DL model and a 2D DL model, both based on the cross-scale attention vision transformer (CrossFormer) neural network, along with a Radiomics model, were developed. These were integrated via ensemble learning into a stacking model. The MultiChannel_2.5D_DL model outperformed the 2D_DL and Radiomics models, with AUCs of 0.806-0.870. The stacking model achieved the highest AUC (0.855-0.904) across validation sets. Patients were stratified into high-risk and low-risk groups based on stacking model scores, with significant survival differences observed via Kaplan-Meier analysis and log-rank tests. The stacking model effectively identifies IDH wt TERTp-mutant gliomas and stratifies patient risk, aiding personalized prognosis.
Collapse
Affiliation(s)
- Wenju Niu
- Department of Radiology, First Hospital of Shanxi Medical University, Taiyuan, 030001, China
- College of Medical Imaging, Shanxi Medical University, Taiyuan, 030001, China
| | - Junyu Yan
- Department of Radiology, First Hospital of Shanxi Medical University, Taiyuan, 030001, China
- Academy of Medical Sciences, Shanxi Medical University, Taiyuan, 030001, China
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, 030001, China
| | - Min Hao
- Department of Radiology, First Hospital of Shanxi Medical University, Taiyuan, 030001, China
- College of Medical Imaging, Shanxi Medical University, Taiyuan, 030001, China
| | - Yibo Zhang
- Faculty of Robotics Science and Engineering, Northeastern University, Shenyang, 110000, China
| | - Tianshi Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100050, China
| | - Chen Liu
- College of Medical Imaging, Shanxi Medical University, Taiyuan, 030001, China
| | - Qijian Li
- College of Medical Imaging, Shanxi Medical University, Taiyuan, 030001, China
| | - Zihao Liu
- College of Medical Imaging, Shanxi Medical University, Taiyuan, 030001, China
| | - Yincheng Su
- College of Medical Imaging, Shanxi Medical University, Taiyuan, 030001, China
| | - Bo Peng
- College of Medical Imaging, Shanxi Medical University, Taiyuan, 030001, China
| | - Yan Tan
- Department of Radiology, First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Xiaochun Wang
- Department of Radiology, First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Lei Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100050, China.
| | - Hui Zhang
- Department of Radiology, First Hospital of Shanxi Medical University, Taiyuan, 030001, China.
- College of Medical Imaging, Shanxi Medical University, Taiyuan, 030001, China.
- Shanxi Key Laboratory of Intelligent Imaging and Nanomedicine, First Hospital of Shanxi Medical University, Taiyuan, 030001, China.
- Intelligent Imaging Big Data and Functional Nano-imaging Engineering Research Center of Shanxi Province, First Hospital of Shanxi Medical University, Taiyuan, 030001, China.
| | - Guoqiang Yang
- Department of Radiology, First Hospital of Shanxi Medical University, Taiyuan, 030001, China.
- College of Medical Imaging, Shanxi Medical University, Taiyuan, 030001, China.
- Shanxi Key Laboratory of Intelligent Imaging and Nanomedicine, First Hospital of Shanxi Medical University, Taiyuan, 030001, China.
| |
Collapse
|
12
|
Cao Y, Liang F, Zhao T, Han J, Wang Y, Wu H, Zhang K, Qiu H, Ding Y, Zhu H. Brain tumor intelligent diagnosis based on Auto-Encoder and U-Net feature extraction. PLoS One 2025; 20:e0315631. [PMID: 40127071 PMCID: PMC11932485 DOI: 10.1371/journal.pone.0315631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 11/27/2024] [Indexed: 03/26/2025] Open
Abstract
Preoperative classification of brain tumors is critical to developing personalized treatment plans, however existing classification methods rely on manual intervention and often have problems with efficiency and accuracy, which may lead to misdiagnosis or delayed diagnosis in clinical practice and affect the therapeutic effect. We propose a fully automated approach to brain tumor magnetic resonance imaging (MRI) classification, consisted by a feature extractor based on the improved U-Net and a classifier based on convolutional recurrent neural network (CRNN). The encoder of the feature extractor based on dense block, is used to enhance feature propagation and reduce the number of parameters. The decoder uses residual block to reduce the weight of some features for improving the effect of MRI spatial sequence reconstruction, and avoid gradient disappearance. Skip connections between the encoder and the decoder effectively merge low-level features and high-level features. The extract feature sequence is input into the CRNN-based classifier for final classification. We assessed the performance of our method for grading glioma, glioma isocitrate dehydrogenase1 (IDH1) mutation status classification and pituitary tumor texture classification on two datasets, glioma or pituitary tumors collected in a local affiliated hospital and glioma imaging data from TCIA. Compared with commonly models and new models, our model achieves higher accuracy, with an accuracy of 90.72%, classified glioma IDH1 mutation status with an accuracy of 94.35%, and classified pituitary tumor texture with an accuracy of 94.64%.
Collapse
Affiliation(s)
- Yaru Cao
- School of Medical Information and Engineering, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Fengning Liang
- School of Medical Information and Engineering, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Teng Zhao
- School of Medical Information and Engineering, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jinting Han
- School of Medical Information and Engineering, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yingchao Wang
- School of Medical Information and Engineering, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Haowen Wu
- School of Medical Information and Engineering, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Kexing Zhang
- School of Medical Information and Engineering, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Huiwen Qiu
- School of Medical Information and Engineering, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yizhe Ding
- School of Medical Information and Engineering, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Hong Zhu
- School of Medical Information and Engineering, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Computer Science and Engineering, State University of New York at Buffalo, Buffalo, New York, United States of America
| |
Collapse
|
13
|
He J, Li H, Zhang B, Liang G, Zhang L, Zhao W, Zhao W, Zhang YJ, Wang ZX, Li JF. Convolutional neural network-assisted Raman spectroscopy for high-precision diagnosis of glioblastoma. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2025; 329:125615. [PMID: 39721487 DOI: 10.1016/j.saa.2024.125615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 12/10/2024] [Accepted: 12/16/2024] [Indexed: 12/28/2024]
Abstract
Glioblastoma multiforme (GBM) is the most lethal intracranial tumor with a median survival of approximately 15 months. Due to its highly invasive properties, it is particularly difficult to accurately identify the tumor margins intraoperatively. The current gold standard for diagnosing GBM during surgery is pathology, but it is time-consuming. Under these circumstances, we developed a method combining Raman spectroscopy (RS) with convolutional neural networks (CNN) to distinguish GBM. Analysis of the spectra of normal brain samples (478 spectra) and GBM samples (462 spectra) from 29 in situ intracranial tumor-bearing mice showed that this method identified GBM tissue with 96.8 % accuracy. Subsequently, spectral analysis of 23 normal human brain tissues (223 spectra) versus 21 tissues from patients with pathologically diagnosed GBM (267 spectra) revealed that the accuracy of this method was 93.9 %. Most importantly, for the difference peaks in the spectra of GBM and normal brain tissue, the common difference peaks in the mouse and human spectra were at 750 cm-1, 1440 cm-1, and 1586 cm-1, which emphasized the differences in cytochrome C and lipids between GBM samples and normal brain samples in both mice and human. The preliminary results showed that CNN-assisted RS is simple to operate and can rapidly and accurately identify whether it is GBM tissue or normal brain tissue.
Collapse
Affiliation(s)
- Jiawei He
- Department of Neurosurgery and Department of Neuroscience, The First Affiliated Hospital of Xiamen University, School of Medicine, College of Chemistry and Chemical Engineering, College of Energy, Institute of Artificial Intelligence, State Key Laboratory for Physical Chemistry of Solid Surfaces, Xiamen University, Xiamen 361102, China
| | - Hongmei Li
- Department of Neurosurgery and Department of Neuroscience, The First Affiliated Hospital of Xiamen University, School of Medicine, College of Chemistry and Chemical Engineering, College of Energy, Institute of Artificial Intelligence, State Key Laboratory for Physical Chemistry of Solid Surfaces, Xiamen University, Xiamen 361102, China
| | - Bingchang Zhang
- Department of Neurosurgery and Department of Neuroscience, The First Affiliated Hospital of Xiamen University, School of Medicine, College of Chemistry and Chemical Engineering, College of Energy, Institute of Artificial Intelligence, State Key Laboratory for Physical Chemistry of Solid Surfaces, Xiamen University, Xiamen 361102, China
| | - Gehao Liang
- Department of Neurosurgery and Department of Neuroscience, The First Affiliated Hospital of Xiamen University, School of Medicine, College of Chemistry and Chemical Engineering, College of Energy, Institute of Artificial Intelligence, State Key Laboratory for Physical Chemistry of Solid Surfaces, Xiamen University, Xiamen 361102, China
| | - Liang Zhang
- Department of Neurosurgery and Department of Neuroscience, The First Affiliated Hospital of Xiamen University, School of Medicine, College of Chemistry and Chemical Engineering, College of Energy, Institute of Artificial Intelligence, State Key Laboratory for Physical Chemistry of Solid Surfaces, Xiamen University, Xiamen 361102, China
| | - Wentao Zhao
- Department of Neurosurgery and Department of Neuroscience, The First Affiliated Hospital of Xiamen University, School of Medicine, College of Chemistry and Chemical Engineering, College of Energy, Institute of Artificial Intelligence, State Key Laboratory for Physical Chemistry of Solid Surfaces, Xiamen University, Xiamen 361102, China
| | - Wenpeng Zhao
- Department of Neurosurgery and Department of Neuroscience, The First Affiliated Hospital of Xiamen University, School of Medicine, College of Chemistry and Chemical Engineering, College of Energy, Institute of Artificial Intelligence, State Key Laboratory for Physical Chemistry of Solid Surfaces, Xiamen University, Xiamen 361102, China
| | - Yue-Jiao Zhang
- Department of Neurosurgery and Department of Neuroscience, The First Affiliated Hospital of Xiamen University, School of Medicine, College of Chemistry and Chemical Engineering, College of Energy, Institute of Artificial Intelligence, State Key Laboratory for Physical Chemistry of Solid Surfaces, Xiamen University, Xiamen 361102, China.
| | - Zhan-Xiang Wang
- Department of Neurosurgery and Department of Neuroscience, The First Affiliated Hospital of Xiamen University, School of Medicine, College of Chemistry and Chemical Engineering, College of Energy, Institute of Artificial Intelligence, State Key Laboratory for Physical Chemistry of Solid Surfaces, Xiamen University, Xiamen 361102, China.
| | - Jian-Feng Li
- Department of Neurosurgery and Department of Neuroscience, The First Affiliated Hospital of Xiamen University, School of Medicine, College of Chemistry and Chemical Engineering, College of Energy, Institute of Artificial Intelligence, State Key Laboratory for Physical Chemistry of Solid Surfaces, Xiamen University, Xiamen 361102, China; Scientific Research Foundation of State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiamen 361005, China.
| |
Collapse
|
14
|
Byeon Y, Park YW, Lee S, Park D, Shin H, Han K, Chang JH, Kim SH, Lee SK, Ahn SS, Hwang D. Interpretable multimodal transformer for prediction of molecular subtypes and grades in adult-type diffuse gliomas. NPJ Digit Med 2025; 8:140. [PMID: 40044878 PMCID: PMC11883078 DOI: 10.1038/s41746-025-01530-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 02/19/2025] [Indexed: 03/09/2025] Open
Abstract
Molecular subtyping and grading of adult-type diffuse gliomas are essential for treatment decisions and patient prognosis. We introduce GlioMT, an interpretable multimodal transformer that integrates imaging and clinical data to predict the molecular subtype and grade of adult-type diffuse gliomas according to the 2021 WHO classification. GlioMT is trained on multiparametric MRI data from an institutional set of 1053 patients with adult-type diffuse gliomas to predict the IDH mutation status, 1p/19q codeletion status, and tumor grade. External validation on the TCGA (200 patients) and UCSF (477 patients) shows that GlioMT outperforms conventional CNNs and visual transformers, achieving AUCs of 0.915 (TCGA) and 0.981 (UCSF) for IDH mutation, 0.854 (TCGA) and 0.806 (UCSF) for 1p/19q codeletion, and 0.862 (TCGA) and 0.960 (UCSF) for grade prediction. GlioMT enhances the reliability of clinical decision-making by offering interpretability through attention maps and contributions of imaging and clinical data.
Collapse
Affiliation(s)
- Yunsu Byeon
- School of Electrical and Electronic Engineering, Yonsei University, Seoul, Republic of 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, Republic of Korea
| | - Soohyun Lee
- School of Electrical and Electronic Engineering, Yonsei University, Seoul, Republic of Korea
| | - Doohyun Park
- School of Electrical and Electronic Engineering, Yonsei University, Seoul, Republic of Korea
| | - HyungSeob Shin
- School of Electrical and Electronic Engineering, Yonsei University, Seoul, Republic of Korea
| | - Kyunghwa Han
- Department of Radiology and Research Institute of Radiological Science and Center for Clinical Imaging Data Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jong Hee Chang
- Department of Neurosurgery, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Se Hoon Kim
- Department of Pathology, Yonsei University College of Medicine, Seoul, Republic of 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, Republic of 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, Republic of Korea.
| | - Dosik Hwang
- School of Electrical and Electronic Engineering, Yonsei University, Seoul, Republic of Korea.
- Department of Radiology and Research Institute of Radiological Science and Center for Clinical Imaging Data Science, Yonsei University College of Medicine, Seoul, Republic of Korea.
- Artificial Intelligence and Robotics Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea.
- Department of Oral and Maxillofacial Radiology, Yonsei University College of Dentistry, Seoul, Republic of Korea.
| |
Collapse
|
15
|
Fathi Kazerooni A, Akbari H, Hu X, Bommineni V, Grigoriadis D, Toorens E, Sako C, Mamourian E, Ballinger D, Sussman R, Singh A, Verginadis II, Dahmane N, Koumenis C, Binder ZA, Bagley SJ, Mohan S, Hatzigeorgiou A, O'Rourke DM, Ganguly T, De S, Bakas S, Nasrallah MP, Davatzikos C. The radiogenomic and spatiogenomic landscapes of glioblastoma and their relationship to oncogenic drivers. COMMUNICATIONS MEDICINE 2025; 5:55. [PMID: 40025245 PMCID: PMC11873127 DOI: 10.1038/s43856-025-00767-0] [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: 09/14/2023] [Accepted: 02/12/2025] [Indexed: 03/04/2025] Open
Abstract
BACKGROUND Glioblastoma is a highly heterogeneous brain tumor, posing challenges for precision therapies and patient stratification in clinical trials. Understanding how genetic mutations influence tumor imaging may improve patient management and treatment outcomes. This study investigates the relationship between imaging features, spatial patterns of tumor location, and genetic alterations in IDH-wildtype glioblastoma, as well as the likely sequence of mutational events. METHODS We conducted a retrospective analysis of 357 IDH-wildtype glioblastomas with pre-operative multiparametric MRI and targeted genetic sequencing data. Radiogenomic signatures and spatial distribution maps were generated for key mutations in genes such as EGFR, PTEN, TP53, and NF1 and their corresponding pathways. Machine and deep learning models were used to identify imaging biomarkers and stratify tumors based on their genetic profiles and molecular heterogeneity. RESULTS Here, we show that glioblastoma mutations produce distinctive imaging signatures, which are more pronounced in tumors with less molecular heterogeneity. These signatures provide insights into how mutations affect tumor characteristics such as neovascularization, cell density, invasion, and vascular leakage. We also found that tumor location and spatial distribution correlate with genetic profiles, revealing associations between tumor regions and specific oncogenic drivers. Additionally, imaging features reflect the cross-sectionally inferred evolutionary trajectories of glioblastomas. CONCLUSIONS This study establishes clinically accessible imaging biomarkers that capture the molecular composition and oncogenic drivers of glioblastoma. These findings have potential implications for noninvasive tumor profiling, personalized therapies, and improved patient stratification in clinical trials.
Collapse
Affiliation(s)
- Anahita Fathi Kazerooni
- AI2D Center for AI and Data Science for Integrated Diagnostics, University of Pennsylvania, Philadelphia, PA, USA
- Center for Data-Driven Discovery in Biomedicine (D3b), Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hamed Akbari
- Department of Bioengineering, School of Engineering, Santa Clara University, Santa Clara, CA, USA
| | - Xiaoju Hu
- Rutgers Cancer Institute of New Jersey, Rutgers the State University of New Jersey, New Brunswick, NJ, USA
| | - Vikas Bommineni
- AI2D Center for AI and Data Science for Integrated Diagnostics, University of Pennsylvania, Philadelphia, PA, USA
| | - Dimitris Grigoriadis
- Department of Computer Science and Biomedical Informatics, University of Thessaly, Lamia, Greece
| | - Erik Toorens
- Penn Genomic Analysis Core, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Chiharu Sako
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth Mamourian
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Dominique Ballinger
- Department of Pathology & Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Robyn Sussman
- Department of Pathology & Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ashish Singh
- AI2D Center for AI and Data Science for Integrated Diagnostics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ioannis I Verginadis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nadia Dahmane
- Department of Neurological Surgery, Weill Cornell Medicine, New York, NY, USA
| | - Constantinos Koumenis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zev A Binder
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Stephen J Bagley
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Suyash Mohan
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Artemis Hatzigeorgiou
- Department of Computer Science and Biomedical Informatics, University of Thessaly, Lamia, Greece
- Hellenic Pasteur Institute, Athens, Greece
| | - Donald M O'Rourke
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Tapan Ganguly
- Penn Genomic Analysis Core, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Subhajyoti De
- Rutgers Cancer Institute of New Jersey, Rutgers the State University of New Jersey, New Brunswick, NJ, USA
| | - Spyridon Bakas
- Department of Pathology & Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - MacLean P Nasrallah
- AI2D Center for AI and Data Science for Integrated Diagnostics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology & Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christos Davatzikos
- AI2D Center for AI and Data Science for Integrated Diagnostics, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
16
|
Xie Y, Zhao C, Zhang X, Shen C, Qi Z, Tang Q, Guo W, Shi Z, Ding H, Yang B, Yu J. Intraoperative Real-Time IDH Diagnosis for Glioma Based on Automatic Analysis of Contrast-Enhanced Ultrasound Video. ULTRASOUND IN MEDICINE & BIOLOGY 2025; 51:484-493. [PMID: 39674714 DOI: 10.1016/j.ultrasmedbio.2024.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 02/15/2024] [Accepted: 11/04/2024] [Indexed: 12/16/2024]
Abstract
OBJECTIVE Isocitrate dehydrogenase (IDH) is the most important molecular marker of glioma and is highly correlated to the diagnosis, treatment, and prognosis of patients. We proposed a real-time diagnosis method for IDH status differentiation based on automatic analysis of intraoperative contrast-enhanced ultrasound (CEUS) video. METHODS Inspired by the Time Intensity Curve (TIC) analysis of CEUS utilized in clinical practice, this paper proposed an automatic CEUS video analysis method called ATAN (Automatic TIC Analysis Network). Based on tumor identification, ATAN automatically selected ROIs (region of interest) inside and outside glioma. ATAN ensures the integrity of dynamic features of perfusion changes at critical locations, resulting in optimal diagnostic performance. The transfer learning mechanism was also introduced by using two auxiliary CEUS datasets to solve the small sample problem of intraoperative glioma data. RESULTS Through pretraining on 258 patients on two auxiliary cohorts, ATAN produced the IDH diagnosis with accuracy and AUC of 0.9 and 0.91 respectively on the main cohort of 60 glioma patients (mean age, 50 years ± 14, 28 men) Compared with other existing IDH status differentiation methods, ATAN is a real-time IDH diagnosis method without the need of tumor samples. CONCLUSION ATAN is an effective automatic analysis model of CEUS, with the help of this model, real-time intraoperative diagnosis of IDH with high accuracy can be achieved. Compared with other state-of-the-art deep learning methods, the accuracy of the ATAN model is 15% higher on average.
Collapse
Affiliation(s)
- Yuanxin Xie
- School of Information Science and Technology, Fudan University, Shanghai, China; Neurosurgical Institute of Fudan University, Shanghai, China
| | - Chengqian Zhao
- School of Information Science and Technology, Fudan University, Shanghai, China; Neurosurgical Institute of Fudan University, Shanghai, China
| | - Xiandi Zhang
- Department of Ultrasound, Huashan Hospital, Fudan University, Shanghai, China
| | - Chao Shen
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China; National Center for Neurological Disorders, Shanghai, China; Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China; Neurosurgical Institute of Fudan University, Shanghai, China
| | - Zengxin Qi
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China; National Center for Neurological Disorders, Shanghai, China; Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China; Neurosurgical Institute of Fudan University, Shanghai, China
| | - Qisheng Tang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China; National Center for Neurological Disorders, Shanghai, China; Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China; Neurosurgical Institute of Fudan University, Shanghai, China
| | - Wei Guo
- Research Institute, VINNO Technology (Suzhou)Co., Ltd., Suzhou, Jiangsu, China
| | - Zhifeng Shi
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China; National Center for Neurological Disorders, Shanghai, China; Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China; Neurosurgical Institute of Fudan University, Shanghai, China
| | - Hong Ding
- Department of Ultrasound, Huashan Hospital, Fudan University, Shanghai, China
| | - Bojie Yang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China; National Center for Neurological Disorders, Shanghai, China; Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China; Neurosurgical Institute of Fudan University, Shanghai, China.
| | - Jinhua Yu
- School of Information Science and Technology, Fudan University, Shanghai, China; Neurosurgical Institute of Fudan University, Shanghai, China
| |
Collapse
|
17
|
Wang Z, Wang L, Wang Y. Radiomics in glioma: emerging trends and challenges. Ann Clin Transl Neurol 2025; 12:460-477. [PMID: 39901654 PMCID: PMC11920724 DOI: 10.1002/acn3.52306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 12/11/2024] [Accepted: 12/31/2024] [Indexed: 02/05/2025] Open
Abstract
Radiomics is a promising neuroimaging technique for extracting and analyzing quantitative glioma features. This review discusses the application, emerging trends, and challenges associated with using radiomics in glioma. Integrating deep learning algorithms enhances various radiomics components, including image normalization, region of interest segmentation, feature extraction, feature selection, and model construction and can potentially improve model accuracy and performance. Moreover, investigating specific tumor habitats of glioblastomas aids in a better understanding of glioblastoma aggressiveness and the development of effective treatment strategies. Additionally, advanced imaging techniques, such as diffusion-weighted imaging, perfusion-weighted imaging, magnetic resonance spectroscopy, magnetic resonance fingerprinting, functional MRI, and positron emission tomography, can provide supplementary information for tumor characterization and classification. Furthermore, radiomics analysis helps understand the glioma immune microenvironment by predicting immune-related biomarkers and characterizing immune responses within tumors. Integrating multi-omics data, such as genomics, transcriptomics, proteomics, and pathomics, with radiomics, aids the understanding of the biological significance of the underlying radiomics features and improves the prediction of genetic mutations, prognosis, and treatment response in patients with glioma. Addressing challenges, such as model reproducibility, model generalizability, model interpretability, and multi-omics data integration, is crucial for the clinical translation of radiomics in glioma.
Collapse
Affiliation(s)
- Zihan Wang
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Lei Wang
- Department of NeurosurgeryGuiqian International General HospitalGuiyangGuizhouChina
| | - Yinyan Wang
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|
18
|
Wang H, Hong Y, Zhang Z, Cheng K, Chen B, Zhang R. Study on postoperative survival prediction model for non-small cell lung cancer: application of radiomics technology workflow based on multi-organ imaging features and various machine learning algorithms. Front Med (Lausanne) 2025; 12:1517765. [PMID: 39975681 PMCID: PMC11835680 DOI: 10.3389/fmed.2025.1517765] [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: 10/27/2024] [Accepted: 01/24/2025] [Indexed: 02/21/2025] Open
Abstract
Objective This study aims to construct an effective prediction model for the two-year postoperative survival probability of patients with non-small cell lung cancer (NSCLC). It particularly focuses on integrating radiomics features, including the erector spinae and whole-lung imaging features, to enhance the accuracy and stability of prognostic predictions. Materials and methods The study included 37 NSCLC patients diagnosed and surgically treated at the First Affiliated Hospital of Anhui Medical University from January 2020 to December 2021. The average age of the patients was 59 years, with the majority being female and non-smokers. Additionally, CT imaging data from 98 patients were obtained from The Cancer Imaging Archive (TCIA) public database. All imaging data were derived from preoperative chest CT scans and standardized using 3D Slicer software. The study extracted radiomic features from the tumor, whole lung, and erector spinae muscles of the patients and applied 11 machine learning algorithms to construct prediction models. Subsequently, the classification performance of all constructed models was compared to select the optimal prediction model. Results Univariate Cox regression analysis showed no significant correlation between the collected clinical factors and patient survival time. In the external validation set, the K-Nearest Neighbors (KNN) model based on bilateral erector spinae features performed the best, with accuracy and AUC (Area Under the Curve) values consistently above 0.7 in both the training and external testing sets. Among the prognostic models based on whole-lung imaging features, the AdaBoost model also performed well, but its AUC value was below 0.6 in the external validation set, indicating overall classification performance still inferior to the KNN model based on erector spinae features. Conclusion This study is the first to introduce erector spinae imaging features into lung cancer research, successfully developing a stable and well-performing prediction model for the postoperative survival of NSCLC patients. The research results provide new perspectives and directions for the application of radiomics in cancer research and emphasize the importance of incorporating multi-organ imaging features to improve the accuracy and stability of prediction models.
Collapse
Affiliation(s)
- Hanlin Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yuan Hong
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zimo Zhang
- Department of The First Clinical Medical College, Anhui Medical University, Hefei, China
| | - Kang Cheng
- Department of The First Clinical Medical College, Anhui Medical University, Hefei, China
| | - Bo Chen
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Renquan Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
19
|
Jin J, Zhang J, Yu X, Xiang Z, Zhu X, Guo M, Zhao Z, Li W, Li H, Xu J, Jin X. Radiomics-guided generative adversarial network for automatic primary target volume segmentation for nasopharyngeal carcinoma using computed tomography images. Med Phys 2025; 52:1119-1132. [PMID: 39535436 DOI: 10.1002/mp.17493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Automatic primary gross tumor volume (GTVp) segmentation for nasopharyngeal carcinoma (NPC) is a quite challenging task because of the existence of similar visual characteristics between tumors and their surroundings, especially on computed tomography (CT) images with severe low contrast resolution. Therefore, most recently proposed methods based on radiomics or deep learning (DL) is difficult to achieve good results on CT datasets. PURPOSE A peritumoral radiomics-guided generative adversarial network (PRG-GAN) was proposed to address this challenge. METHODS A total of 157 NPC patients with CT images was collected and divided into training, validation, and testing cohorts of 108, 9, and 30 patients, respectively. The proposed model was based on a standard GAN consisting of a generator network and a discriminator network. Morphological dilation on the initial segmentation results from GAN was first conducted to delineate annular peritumoral region, in which radiomics features were extracted as priori guide knowledge. Then, radiomics features were fused with semantic features by the discriminator's fully connected layer to achieve the voxel-level classification and segmentation. The dice similarity coefficient (DSC), 95% Hausdorff distance (HD95), and average symmetric surface distance (ASSD) were used to evaluate the segmentation performance using a paired samples t-test with Bonferroni correction and Cohen's d (d) effect sizes. A two-sided p-value of less than 0.05 was considered statistically significant. RESULTS The model-generated predictions had a high overlap ratio with the ground truth. The average DSC, HD95, and ASSD were significantly improved from 0.80 ± 0.12, 4.65 ± 4.71 mm, and 1.35 ± 1.15 mm of GAN to 0.85 ± 0.18 (p = 0.001, d = 0.71), 4.15 ± 7.56 mm (p = 0.002, d = 0.67), and 1.11 ± 1.65 mm (p < 0.001, d = 0.46) of PRG-GAN, respectively. CONCLUSION Integrating radiomics features into GAN is promising to solve unclear border limitations and increase the delineation accuracy of GTVp for patients with NPC.
Collapse
Affiliation(s)
- Juebin Jin
- Department of Radiotherapy Center, 1st Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jicheng Zhang
- Department of Radiotherapy Center, 1st Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xianwen Yu
- Department of Radiotherapy Center, 1st Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ziqing Xiang
- Department of Medical Engineering, 2nd Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xuanxuan Zhu
- Department of Radiotherapy Center, 1st Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Mingrou Guo
- Department of Radiotherapy Center, 1st Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zeshuo Zhao
- Department of Radiotherapy Center, 1st Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - WenLong Li
- Department of Radiotherapy Center, 1st Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Heng Li
- Department of Radiotherapy Center, 1st Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jiayi Xu
- Department of Radiotherapy Center, 1st Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiance Jin
- Department of Radiotherapy Center, 1st Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- School of Basic Medical Science, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
20
|
Zhu Y, Wang J, Xue C, Zhai X, Xiao C, Lu T. Deep Learning and Habitat Radiomics for the Prediction of Glioma Pathology Using Multiparametric MRI: A Multicenter Study. Acad Radiol 2025; 32:963-975. [PMID: 39322536 DOI: 10.1016/j.acra.2024.09.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 09/05/2024] [Accepted: 09/06/2024] [Indexed: 09/27/2024]
Abstract
RATIONALE AND OBJECTIVES Recent radiomics studies on predicting pathological outcomes of glioma have shown immense potential. However, the predictive ability remains suboptimal due to the tumor intrinsic heterogeneity. We aimed to achieve better pathological prediction outcomes by combining habitat analysis with deep learning. MATERIALS AND METHODS 387 cases of primary glioma from three hospitals were collected, along with their T1 contrast-enhanced and T2-weighted MR sequences, pathological reports and clinical histories. The training set consisted of 264 patients, 82 patients composed the test set, and 41 patients were used as the validation set for hyperparameter tuning and optimal model selection. All groups were sourced from different centers. Through radiomics, deep learning, habitat analysis and combined analysis, we extracted imaging features separately and jointly modeled them with clinical features. We identified the optimal models for predicting glioma grades, Ki67 expression levels, P53 mutation and IDH1 mutation. RESULTS Using a LightGBM model with DenseNet161 features based on habitat subregions, the best tumor grade prediction model was achieved. A LightGBM model with ResNet50 features based on habitat subregions yielded the best Ki67 expression level prediction model. An SVM model with Radiomics and Inception_v3 features provided the best prediction of P53 mutation. The best model for predicting IDH1 mutation was achieved by an MLP model with Radiomics features based on habitat subregions. Clinical features might be potentially helpful for the prediction with relatively weak evidence. CONCLUSION Habitat+Deep Learning feature extraction methods were optimal for predicting grades and Ki67 levels. Deep Learning is optimal for predicting P53 mutation, while the combination of Habitat+ Radiomics models yielded the best prediction for IDH1 mutation.
Collapse
Affiliation(s)
- Yunyang Zhu
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, China (Y.Z., J.W., T.L.)
| | - Jing Wang
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, China (Y.Z., J.W., T.L.)
| | - Chen Xue
- Department of Radiology, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China (C.X., C.X.)
| | - Xiaoyang Zhai
- The First Affiliated Hospital of Xinxiang University, Xinxiang, China (X.Z.)
| | - Chaoyong Xiao
- Department of Radiology, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China (C.X., C.X.)
| | - Ting Lu
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, China (Y.Z., J.W., T.L.).
| |
Collapse
|
21
|
Wang Y, Gao A, Yang H, Bai J, Zhao G, Zhang H, Song Y, Wang C, Zhang Y, Cheng J, Yang G. Using partially shared radiomics features to simultaneously identify isocitrate dehydrogenase mutation status and epilepsy in glioma patients from MRI images. Sci Rep 2025; 15:3591. [PMID: 39875517 PMCID: PMC11775202 DOI: 10.1038/s41598-025-87778-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 01/22/2025] [Indexed: 01/30/2025] Open
Abstract
Prediction of isocitrate dehydrogenase (IDH) mutation status and epilepsy occurrence are important to glioma patients. Although machine learning models have been constructed for both issues, the correlation between them has not been explored. Our study aimed to exploit this correlation to improve the performance of both of the IDH mutation status identification and epilepsy diagnosis models in patients with glioma II-IV. 399 patients were retrospectively enrolled and divided into a training (n = 279) and an independent test (n = 120) cohort. Multi-center dataset (n = 228) from The Cancer Imaging Archive (TCIA) was used for external test for identification of IDH mutation status. Region of interests comprising the entire tumor and peritumoral edema were automatically segmented using a pre-trained deep learning model. Radiomic features were extracted from T1-weighted, T2-weighted, post-Gadolinium T1 weighted, and T2 fluid-attenuated inversion recovery images. We proposed an iterative approach derived from LASSO to select features shared by two tasks and features specific to each task, before using them to construct the final models. Receiver operating characteristic (ROC) analysis was employed to evaluate the model. The IDH mutation identification model achieved area under the ROC curve (AUC) values of 0.948, 0.946 and 0.860 on the training, internal test, and external test cohorts, respectively. The epilepsy diagnosis model achieved AUCs of 0.924 and 0.880 on the training and internal test cohorts, respectively. The proposed models can identify IDH status and epilepsy with fewer features, thus having better interpretability and lower risk of overfitting. This not only improves its chance of application in clinical settings, but also provides a new scheme to study multiple correlated clinical tasks.
Collapse
Affiliation(s)
- Yida Wang
- Shanghai Key Laboratory of Magnetic Resonance, School of Physics and Electronic Science, East China Normal University, Shanghai, China
- Institute of Magnetic Resonance and Molecular Imaging in Medicine, East China Normal University, Shanghai, China
| | - Ankang Gao
- Department of MRI, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongxi Yang
- Shanghai Key Laboratory of Magnetic Resonance, School of Physics and Electronic Science, East China Normal University, Shanghai, China
- Institute of Magnetic Resonance and Molecular Imaging in Medicine, East China Normal University, Shanghai, China
| | - Jie Bai
- Department of MRI, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guohua Zhao
- Department of MRI, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Huiting Zhang
- MR Scientific Marketing, Siemens Healthineers China, Shanghai, China
| | - Yang Song
- MR Scientific Marketing, Siemens Healthineers China, Shanghai, China
| | - Chenglong Wang
- Shanghai Key Laboratory of Magnetic Resonance, School of Physics and Electronic Science, East China Normal University, Shanghai, China
- Institute of Magnetic Resonance and Molecular Imaging in Medicine, East China Normal University, Shanghai, China
| | - Yong Zhang
- Department of MRI, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jingliang Cheng
- Department of MRI, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Guang Yang
- Shanghai Key Laboratory of Magnetic Resonance, School of Physics and Electronic Science, East China Normal University, Shanghai, China.
- Institute of Magnetic Resonance and Molecular Imaging in Medicine, East China Normal University, Shanghai, China.
| |
Collapse
|
22
|
Dorfner FJ, Patel JB, Kalpathy-Cramer J, Gerstner ER, Bridge CP. A review of deep learning for brain tumor analysis in MRI. NPJ Precis Oncol 2025; 9:2. [PMID: 39753730 PMCID: PMC11698745 DOI: 10.1038/s41698-024-00789-2] [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: 04/23/2024] [Accepted: 12/17/2024] [Indexed: 01/06/2025] Open
Abstract
Recent progress in deep learning (DL) is producing a new generation of tools across numerous clinical applications. Within the analysis of brain tumors in magnetic resonance imaging, DL finds applications in tumor segmentation, quantification, and classification. It facilitates objective and reproducible measurements crucial for diagnosis, treatment planning, and disease monitoring. Furthermore, it holds the potential to pave the way for personalized medicine through the prediction of tumor type, grade, genetic mutations, and patient survival outcomes. In this review, we explore the transformative potential of DL for brain tumor care and discuss existing applications, limitations, and future directions and opportunities.
Collapse
Affiliation(s)
- Felix J Dorfner
- Athinoula A. Martinos Center for Biomedical Imaging, 149 13th St, Charlestown, MA, 02129, USA
| | - Jay B Patel
- Athinoula A. Martinos Center for Biomedical Imaging, 149 13th St, Charlestown, MA, 02129, USA
| | | | - Elizabeth R Gerstner
- Athinoula A. Martinos Center for Biomedical Imaging, 149 13th St, Charlestown, MA, 02129, USA
- Massachusetts General Hospital Cancer Center, Boston, MA, 02114, USA
| | - Christopher P Bridge
- Athinoula A. Martinos Center for Biomedical Imaging, 149 13th St, Charlestown, MA, 02129, USA.
| |
Collapse
|
23
|
Foltyn-Dumitru M, Rastogi A, Cho J, Schell M, Mahmutoglu MA, Kessler T, Sahm F, Wick W, Bendszus M, Brugnara G, Vollmuth P. The potential of GPT-4 advanced data analysis for radiomics-based machine learning models. Neurooncol Adv 2025; 7:vdae230. [PMID: 39780768 PMCID: PMC11707530 DOI: 10.1093/noajnl/vdae230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025] Open
Abstract
Background This study aimed to explore the potential of the Advanced Data Analytics (ADA) package of GPT-4 to autonomously develop machine learning models (MLMs) for predicting glioma molecular types using radiomics from MRI. Methods Radiomic features were extracted from preoperative MRI of n = 615 newly diagnosed glioma patients to predict glioma molecular types (IDH-wildtype vs IDH-mutant 1p19q-codeleted vs IDH-mutant 1p19q-non-codeleted) with a multiclass ML approach. Specifically, ADA was used to autonomously develop an ML pipeline and benchmark performance against an established handcrafted model using various MRI normalization methods (N4, Zscore, and WhiteStripe). External validation was performed on 2 public glioma datasets D2 (n = 160) and D3 (n = 410). Results GPT-4 achieved the highest accuracy of 0.820 (95% CI = 0.819-0.821) on the D3 dataset with N4/WS normalization, significantly outperforming the benchmark model's accuracy of 0.678 (95% CI = 0.677-0.680) (P < .001). Class-wise analysis showed performance variations across different glioma types. In the IDH-wildtype group, GPT-4 had a recall of 0.997 (95% CI = 0.997-0.997), surpassing the benchmark's 0.742 (95% CI = 0.740-0.743). For the IDH-mut 1p/19q-non-codel group, GPT-4's recall was 0.275 (95% CI = 0.272-0.279), lower than the benchmark's 0.426 (95% CI = 0.423-0.430). In the IDH-mut 1p/19q-codel group, GPT-4's recall was 0.199 (95% CI = 0.191-0.206), below the benchmark's 0.730 (95% CI = 0.721-0.738). On the D2 dataset, GPT-4's accuracy was significantly lower (P < .001) than the benchmark's, with N4/WS achieving 0.668 (95% CI = 0.666-0.671) compared with 0.719 (95% CI = 0.717-0.722) (P < .001). Class-wise analysis revealed the same pattern as observed in D3. Conclusions GPT-4 can autonomously develop radiomics-based MLMs, achieving performance comparable to handcrafted MLMs. However, its poorer class-wise performance due to unbalanced datasets shows limitations in handling complete end-to-end ML pipelines.
Collapse
Affiliation(s)
- Martha Foltyn-Dumitru
- Division for Computational Radiology & Clinical AI (CCIBonn.ai), Department of Neuroradiology, Bonn University Hospital, Bonn, Germany
- Division for Computational Neuroimaging, Heidelberg University Hospital, Heidelberg, Germany
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Aditya Rastogi
- Division for Computational Radiology & Clinical AI (CCIBonn.ai), Department of Neuroradiology, Bonn University Hospital, Bonn, Germany
- Division for Computational Neuroimaging, Heidelberg University Hospital, Heidelberg, Germany
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Jaeyoung Cho
- Division for Computational Radiology & Clinical AI (CCIBonn.ai), Department of Neuroradiology, Bonn University Hospital, Bonn, Germany
- Division for Computational Neuroimaging, Heidelberg University Hospital, Heidelberg, Germany
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Marianne Schell
- Division for Computational Neuroimaging, Heidelberg University Hospital, Heidelberg, Germany
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Mustafa Ahmed Mahmutoglu
- Division for Computational Neuroimaging, Heidelberg University Hospital, Heidelberg, Germany
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Tobias Kessler
- Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neurology and Neurooncology Program, Heidelberg University Hospital, Heidelberg University, Heidelberg, Germany
| | - Felix Sahm
- Clinical Cooperation Unit Neuropathology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neuropathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Wolfgang Wick
- Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neurology and Neurooncology Program, Heidelberg University Hospital, Heidelberg University, Heidelberg, Germany
| | - Martin Bendszus
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Gianluca Brugnara
- Division for Medical Image Computing (MIC), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division for Computational Radiology & Clinical AI (CCIBonn.ai), Department of Neuroradiology, Bonn University Hospital, Bonn, Germany
- Division for Computational Neuroimaging, Heidelberg University Hospital, Heidelberg, Germany
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Philipp Vollmuth
- Division for Medical Image Computing (MIC), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division for Computational Radiology & Clinical AI (CCIBonn.ai), Department of Neuroradiology, Bonn University Hospital, Bonn, Germany
- Division for Computational Neuroimaging, Heidelberg University Hospital, Heidelberg, Germany
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
24
|
Xiao ML, Fu L, Qian T, Wei Y, Ma FH, Li YA, Cheng JJ, Qian ZX, Zhang GF, Qiang JW. The deep learning radiomics nomogram helps to evaluate the lymph node status in cervical adenocarcinoma/adenosquamous carcinoma. Front Oncol 2024; 14:1414609. [PMID: 39735600 PMCID: PMC11671353 DOI: 10.3389/fonc.2024.1414609] [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: 04/15/2024] [Accepted: 11/20/2024] [Indexed: 12/31/2024] Open
Abstract
Objectives The accurate assessment of lymph node metastasis (LNM) can facilitate clinical decision-making on radiotherapy or radical hysterectomy (RH) in cervical adenocarcinoma (AC)/adenosquamous carcinoma (ASC). This study aims to develop a deep learning radiomics nomogram (DLRN) to preoperatively evaluate LNM in cervical AC/ASC. Materials and methods A total of 652 patients from a multicenter were enrolled and randomly allocated into primary, internal, and external validation cohorts. The radiomics features were extracted from axial T2-weighted imaging (T2WI), diffusion-weighted imaging (DWI), and contrast-enhanced T1-weighted imaging (CE-T1WI). The DL features from T2WI, DWI, and CE-T1WI were exported from Resnet 34, which was pretrained by 14 million natural images of the ImageNet dataset. The radscore (RS) and DL score (DLS) were independently obtained after repeatability test, Pearson correlation coefficient (PCC), minimum redundancy maximum relevance (MRMR), and least absolute shrinkage and selection operator (LASSO) algorithm performed on the radiomics and DL feature sets. The DLRN was then developed by integrating the RS, DLS, and independent clinicopathological factors for evaluating the LNM in cervical AC/ASC. Results The nomogram of DLRN-integrated FIGO stage, menopause, RS, and DLS achieved AUCs of 0.79 (95% CI, 0.74-0.83), 0.87 (95% CI, 0.81-0.92), and 0.86 (95% CI, 0.79-0.91) in the primary, internal, and external validation cohorts. Compared with the RS, DLS, and clinical models, DLRN had a significant higher AUC for evaluating LNM (all P < 0.005). Conclusions The nomogram of DLRN can accurately evaluate LNM in cervical AC/ASC.
Collapse
Affiliation(s)
- Mei Ling Xiao
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Department of Radiology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Le Fu
- Department of Radiology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ting Qian
- Department of Radiology, International Peace Maternity and Child Health Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yan Wei
- College of Information Engineering, Zhejiang University of Technology, Hangzhou, Zhejiang, China
| | - Feng Hua Ma
- Departments of Radiology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Yong Ai Li
- Department of Radiology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Jie Jun Cheng
- Department of Radiology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhao Xia Qian
- Department of Radiology, International Peace Maternity and Child Health Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Guo Fu Zhang
- Departments of Radiology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Jin Wei Qiang
- Department of Radiology, Jinshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
25
|
Sun C, Jiang C, Wang X, Ma S, Zhang D, Jia W. MR-Based Radiomics Predicts CDK6 Expression and Prognostic Value in High-grade Glioma. Acad Radiol 2024; 31:5141-5153. [PMID: 38964985 DOI: 10.1016/j.acra.2024.06.006] [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: 02/28/2024] [Revised: 05/28/2024] [Accepted: 06/03/2024] [Indexed: 07/06/2024]
Abstract
RATIONALE AND OBJECTIVES This study aims to assess the prognostic value of Cyclin-dependent kinases 6 (CDK6) expression levels and establish a machine learning-based radiomics model for predicting the expression levels of CDK6 in high-grade gliomas (HGG). MATERIALS AND METHODS Clinical parameters and genomic data were extracted from 310 HGG patients in the Cancer Genome Atlas (TCGA) database and 27 patients in the Repository of Molecular Brain Neoplasia Data (REMBRANDT) database. Univariate and multivariate Cox regression, as well as Kaplan-Meier analysis, were performed for prognosis analysis. The correlation between immune cell Infiltration with CDK6 was assessed using spearman correlation analysis. Radiomic features were extracted from contrast-enhanced magnetic resonance imaging (CE-MRI) in the Cancer Imaging Archive (TCIA) database (n = 82) and REMBRANDT database (n = 27). Logistic regression (LR) and support vector machine (SVM) were employed to establish the radiomics model for predicting CDK6 expression. Receiver operating characteristic curve (ROC), calibration curve, and decision curve analysis (DCA) were utilized to assess the predictive performance of the radiomics model. Generate radiomic scores (RS) based on the LR model. An RS-based nomogram was constructed to predict the prognosis of HGG. RESULTS CDK6 was significantly overexpressed in HGG tissues and was related to lower overall survival. A significant elevation in infiltrating M0 macrophages was observed in the CDK6 high group (P < 0.001). The LR radiomics model for the prediction of CDK6 expression levels (AUC=0.810 in the training cohort, AUC = 0.784 after cross-validation, AUC=0.750 in the testing cohort) was established utilizing three radiomic features. The predictive efficiencies of the RS-based nomogram, as measured by AUC, were 0.769 for 1-year, 0.815 for 3-year, and 0.780 for 5-year, respectively. CONCLUSION The expression level of CDK6 can impact the prognosis of patients with HGG. The expression level of HGG can be noninvasively prognosticated utilizing a radiomics model.
Collapse
Affiliation(s)
- Chen Sun
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 West Road, South Fourth Ring Road, Beijing, China
| | - Chenggang Jiang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 West Road, South Fourth Ring Road, Beijing, China
| | - Xi Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 West Road, South Fourth Ring Road, Beijing, China
| | - Shunchang Ma
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 West Road, South Fourth Ring Road, Beijing, China
| | - Dainan Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 West Road, South Fourth Ring Road, Beijing, China
| | - Wang Jia
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 West Road, South Fourth Ring Road, Beijing, China.
| |
Collapse
|
26
|
Liu J, Wang P, Zhang H, Guo Y, Tang M, Wang J, Wu N. Current research status of Raman spectroscopy in glioma detection. Photodiagnosis Photodyn Ther 2024; 50:104388. [PMID: 39461488 DOI: 10.1016/j.pdpdt.2024.104388] [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: 08/11/2024] [Revised: 10/05/2024] [Accepted: 10/18/2024] [Indexed: 10/29/2024]
Abstract
Glioma is the most common primary tumor of the nervous system. Conventional diagnostic methods for glioma often involve time-consuming or reliance on externally introduced materials. Consequently, there is an urgent need for rapid and reliable diagnostic techniques. Raman spectroscopy has emerged as a promising tool, offering rapid, accurate, and label-free analysis with high sensitivity and specificity in biomedical applications. In this review, the fundamental principles of Raman spectroscopy have been introduced, and then the progress of applying Raman spectroscopy in biomedical studies has been summarized, including the identification and typing of glioma. The challenges encountered in the clinical application of Raman spectroscopy for glioma have been discussed, and the prospects have also been envisioned.
Collapse
Affiliation(s)
- Jie Liu
- Department of Neurosurgery, Chongqing General Hospital, Chongqing University, Chongqing 401147, China; Chongqing Research Center for Glioma Precision Medicine, Chongqing University, Chongqing 401147, China
| | - Pan Wang
- Department of Neurosurgery, Chongqing General Hospital, Chongqing University, Chongqing 401147, China; Chongqing Research Center for Glioma Precision Medicine, Chongqing University, Chongqing 401147, China
| | - Hua Zhang
- Chongqing Institute of Green and Intelligent Technology, Chongqing University, Chongqing 400714, China
| | - Yuansen Guo
- Chongqing Institute of Green and Intelligent Technology, Chongqing University, Chongqing 400714, China
| | - Mingjie Tang
- Chongqing Institute of Green and Intelligent Technology, Chongqing University, Chongqing 400714, China
| | - Junwei Wang
- Department of Neurosurgery, Chongqing General Hospital, Chongqing University, Chongqing 401147, China; Chongqing Research Center for Glioma Precision Medicine, Chongqing University, Chongqing 401147, China
| | - Nan Wu
- Department of Neurosurgery, Chongqing General Hospital, Chongqing University, Chongqing 401147, China; Chongqing Research Center for Glioma Precision Medicine, Chongqing University, Chongqing 401147, China.
| |
Collapse
|
27
|
Guo J, Xu P, Wu Y, Tao Y, Han C, Lin J, Zhao K, Liu Z, Liu W, Lu C. CroMAM: A Cross-Magnification Attention Feature Fusion Model for Predicting Genetic Status and Survival of Gliomas Using Histological Images. IEEE J Biomed Health Inform 2024; 28:7345-7356. [PMID: 39028591 DOI: 10.1109/jbhi.2024.3431471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/21/2024]
Abstract
Predicting the gene mutation status in whole slide images (WSIs) is crucial for the clinical treatment, cancer management, and research of gliomas. With advancements in CNN and Transformer algorithms, several promising models have been proposed. However, existing studies have paid little attention on fusing multi-magnification information, and the model requires processing all patches from a whole slide image. In this paper, we propose a cross-magnification attention model called CroMAM for predicting the genetic status and survival of gliomas. The CroMAM first utilizes a systematic patch extraction module to sample a subset of representative patches for downstream analysis. Next, the CroMAM applies Swin Transformer to extract local and global features from patches at different magnifications, followed by acquiring high-level features and dependencies among single-magnification patches through the application of a Vision Transformer. Subsequently, the CroMAM exchanges the integrated feature representations of different magnifications and encourage the integrated feature representations to learn the discriminative information from other magnification. Additionally, we design a cross-magnification attention analysis method to examine the effect of cross-magnification attention quantitatively and qualitatively which increases the model's explainability. To validate the performance of the model, we compare the proposed model with other multi-magnification feature fusion models on three tasks in two datasets. Extensive experiments demonstrate that the proposed model achieves state-of-the-art performance in predicting the genetic status and survival of gliomas.
Collapse
|
28
|
Musigmann M, Bilgin M, Bilgin SS, Krähling H, Heindel W, Mannil M. Completely non-invasive prediction of IDH mutation status based on preoperative native CT images. Sci Rep 2024; 14:26763. [PMID: 39501053 PMCID: PMC11538254 DOI: 10.1038/s41598-024-77789-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 10/25/2024] [Indexed: 11/08/2024] Open
Abstract
The isocitrate dehydrogenase (IDH) mutation status is one of the most important markers according to the 2021 WHO classification of CNS tumors. Preoperatively, this information is usually obtained based on invasive biopsies, contrast-enhanced MR images or PET images generated using radioactive tracers. However, the completely non-invasive determination of IDH mutation status using routinely acquired preoperative native CT images has hardly been investigated to date. In our study, we show that radiomics-based machine learning allows to determine IDH mutation status based on preoperative native CT images both with very high accuracy and completely non-invasively. Based on independent test data, we are able to correctly identify 91.1% of cases with an IDH mutation. Our final model, containing only six features, exhibits a high area under the curve of 0.847 and an excellent area under the precision-recall curve of 0.945. In the future, such models may be used for a completely non-invasive prediction of important genetic markers, potentially allowing treating physicians to reduce the number of biopsies and speed up further treatment planning.
Collapse
Affiliation(s)
- Manfred Musigmann
- University Clinic for Radiology, University Münster and University Hospital Münster, Albert- Schweitzer-Campus 1, 48149, Münster, Germany
| | - Melike Bilgin
- University Clinic for Radiology, University Münster and University Hospital Münster, Albert- Schweitzer-Campus 1, 48149, Münster, Germany
| | - Sabriye Sennur Bilgin
- University Clinic for Radiology, University Münster and University Hospital Münster, Albert- Schweitzer-Campus 1, 48149, Münster, Germany
| | - Hermann Krähling
- University Clinic for Radiology, University Münster and University Hospital Münster, Albert- Schweitzer-Campus 1, 48149, Münster, Germany
| | - Walter Heindel
- University Clinic for Radiology, University Münster and University Hospital Münster, Albert- Schweitzer-Campus 1, 48149, Münster, Germany
| | - Manoj Mannil
- University Clinic for Radiology, University Münster and University Hospital Münster, Albert- Schweitzer-Campus 1, 48149, Münster, Germany.
| |
Collapse
|
29
|
Hu C, Fang K, Du Q, Chen J, Wang L, Zhang J, Bai R, Wang Y. Diffusion-weighted MRI precisely predicts telomerase reverse transcriptase promoter mutation status in World Health Organization grade IV gliomas using a residual convolutional neural network. Br J Radiol 2024; 97:1806-1815. [PMID: 39152999 DOI: 10.1093/bjr/tqae146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 06/18/2024] [Accepted: 08/08/2024] [Indexed: 08/19/2024] Open
Abstract
OBJECTIVES Telomerase reverse transcriptase promoter (pTERT) mutation status plays a key role in making decisions and predicting prognoses for patients with World Health Organization (WHO) grade IV glioma. This study was conducted to assess the value of diffusion-weighted imaging (DWI) for predicting pTERT mutation status in WHO grade IV glioma. METHODS MRI data and molecular information were obtained for 266 patients with WHO grade IV glioma at the hospital and divided into training and validation sets. The ratio of training to validation set was approximately 10:3. We trained the same residual convolutional neural network (ResNet) for each MR modality, including structural MRIs (T1-weighted, T2-weighted, and contrast-enhanced T1-weighted) and DWI*, to compare the predictive capacities between DWI and conventional structural MRI. We also explored the effects of different regions of interest on pTERT mutation status prediction outcomes. RESULTS Structural MRI modalities poorly predicted the pTERT mutation status (accuracy = 51%-54%; area under the curve [AUC]=0.545-0.571), whereas DWI combined with its apparent diffusive coefficient maps yielded the best predictive performance (accuracy = 85.2%, AUC = 0.934). Including the radiological and clinical characteristics did not further improve the performance for predicting pTERT mutation status. The entire tumour volume yielded the best prediction performance. CONCLUSIONS DWI technology shows promising potential for predicting pTERT mutations in WHO grade IV glioma and should be included in the MRI protocol for WHO grade IV glioma in clinical practice. ADVANCES IN KNOWLEDGE This is the first large-scale model study to validate the predictive value of DWI for pTERT in WHO grade IV glioma.
Collapse
Affiliation(s)
- Congman Hu
- Department of Neurosurgery, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
- Department of Neurosurgery, Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou 310009, China
- Department of Neurosurgery, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu 322000, China
| | - Ke Fang
- College of Information Science and Electronic Engineering, Zhejiang University, Hangzhou 310020, China
| | - Quan Du
- Department of Neurosurgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Jiarui Chen
- Department of Neurosurgery, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
- Department of Neurosurgery, Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou 310009, China
| | - Lin Wang
- Department of Neurosurgery, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
- Department of Neurosurgery, Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou 310009, China
| | - Jianmin Zhang
- Department of Neurosurgery, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
- Department of Neurosurgery, Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou 310009, China
| | - Ruiliang Bai
- Department of Physical Medicine and Rehabilitation of the Affiliated Sir Run Run Shaw Hospital and Interdisciplinary Institute of Neuroscience and Technology, School of Medicine, Zhejiang University, Hangzhou 310020, China
- Department of Key Laboratory of Biomedical Engineering of Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou 310058, China
| | - Yongjie Wang
- Department of Neurosurgery, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
- Department of Neurosurgery, Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou 310009, China
| |
Collapse
|
30
|
Fang Z, Shu T, Luo P, Shao Y, Lin L, Tu Z, Zhu X, Wu L. The peritumoral edema index and related mechanisms influence the prognosis of GBM patients. Front Oncol 2024; 14:1417208. [PMID: 39534094 PMCID: PMC11554619 DOI: 10.3389/fonc.2024.1417208] [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: 04/14/2024] [Accepted: 10/08/2024] [Indexed: 11/16/2024] Open
Abstract
Background Peritumoral brain edema (PTBE) represents a characteristic phenotype of intracranial gliomas. However, there is a lack of consensus regarding the prognosis and mechanism of PTBE. In this study, clinical imaging data, along with publicly available imaging data, were utilized to assess the prognosis of PTBE in glioblastoma (GBM) patients, and the associated mechanisms were preliminarily analyzed. Methods We investigated relevant imaging features, including edema, in GBM patients using ITK-SNAP imaging segmentation software. Risk factors affecting progression-free survival (PFS) and overall survival (OS) were assessed using a Cox proportional hazard regression model. In addition, the impact of PTBE on PFS and OS was analyzed in clinical GBM patients using the Kaplan-Meier survival analysis method, and the results further validated by combining data from The Cancer Imaging Archive (TCIA) and The Cancer Genome Atlas (TCGA). Finally, functional enrichment analysis based on TCIA and TCGA datasets identified several pathways potentially involved in the mechanism of edema formation. Results This study included a total of 32 clinical GBM patients and 132 GBM patients from public databases. Univariate and multivariate analyses indicated that age and edema index (EI) are independent risk factors for PFS, but not for OS. Kaplan-Meier curves revealed consistent survival analysis results between IE groups among both clinical patients and TCIA and TCGA patients, suggesting a significant effect of PTBE on PFS but not on OS. Furthermore, functional enrichment analysis predicted the involvement of several pathways related mainly to cellular bioenergetics and vasculogenic processes in the mechanism of PTBE formation. While these novel results warrant confirmation in a larger patient cohort, they support good prognostic value for PTBE assessment in GBM. Conclusions Our results indicate that a low EI positively impacts disease control in GBM patients, but this does not entirely translate into an improvement in OS. Multiple genes, signaling pathways, and biological processes may contribute to the formation of peritumoral edema in GBM through cytotoxic and vascular mechanisms.
Collapse
Affiliation(s)
- Zhansheng Fang
- Department of Neurosurgery, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang University, Nanchang, China
- Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang University, Nanchang, China
- Institute of Neuroscience, Nanchang University, Nanchang, China
| | - Ting Shu
- Department of Medical Imaging Center, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Pengxiang Luo
- Department of Neurosurgery, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang University, Nanchang, China
- Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang University, Nanchang, China
- Institute of Neuroscience, Nanchang University, Nanchang, China
| | - Yiqing Shao
- Department of Neurosurgery, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang University, Nanchang, China
- Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang University, Nanchang, China
- Institute of Neuroscience, Nanchang University, Nanchang, China
| | - Li Lin
- Department of Neurosurgery, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang University, Nanchang, China
- Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang University, Nanchang, China
- Institute of Neuroscience, Nanchang University, Nanchang, China
| | - Zewei Tu
- Department of Neurosurgery, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang University, Nanchang, China
- Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang University, Nanchang, China
- Institute of Neuroscience, Nanchang University, Nanchang, China
| | - Xingen Zhu
- Department of Neurosurgery, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang University, Nanchang, China
- Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang University, Nanchang, China
- Institute of Neuroscience, Nanchang University, Nanchang, China
| | - Lei Wu
- Department of Neurosurgery, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang University, Nanchang, China
- Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang University, Nanchang, China
- Institute of Neuroscience, Nanchang University, Nanchang, China
| |
Collapse
|
31
|
Jacome MA, Wu Q, Piña Y, Etame AB. Evolution of Molecular Biomarkers and Precision Molecular Therapeutic Strategies in Glioblastoma. Cancers (Basel) 2024; 16:3635. [PMID: 39518074 PMCID: PMC11544870 DOI: 10.3390/cancers16213635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 10/24/2024] [Accepted: 10/26/2024] [Indexed: 11/16/2024] Open
Abstract
Glioblastoma is the most commonly occurring malignant brain tumor, with a high mortality rate despite current treatments. Its classification has evolved over the years to include not only histopathological features but also molecular findings. Given the heterogeneity of glioblastoma, molecular biomarkers for diagnosis have become essential for initiating treatment with current therapies, while new technologies for detecting specific variations using computational tools are being rapidly developed. Advances in molecular genetics have made possible the creation of tailored therapies based on specific molecular targets, with various degrees of success. This review provides an overview of the latest advances in the fields of histopathology and radiogenomics and the use of molecular markers for management of glioblastoma, as well as the development of new therapies targeting the most common molecular markers. Furthermore, we offer a summary of the results of recent preclinical and clinical trials to recognize the current trends of investigation and understand the possible future directions of molecular targeted therapies in glioblastoma.
Collapse
Affiliation(s)
- Maria A. Jacome
- Departamento de Ciencias Morfológicas Microscópicas, Universidad de Carabobo, Valencia 02001, Venezuela
| | - Qiong Wu
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA; (Q.W.); (Y.P.)
| | - Yolanda Piña
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA; (Q.W.); (Y.P.)
| | - Arnold B. Etame
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA; (Q.W.); (Y.P.)
| |
Collapse
|
32
|
Singh G, Singh A, Bae J, Manjila S, Spektor V, Prasanna P, Lignelli A. -New frontiers in domain-inspired radiomics and radiogenomics: increasing role of molecular diagnostics in CNS tumor classification and grading following WHO CNS-5 updates. Cancer Imaging 2024; 24:133. [PMID: 39375809 PMCID: PMC11460168 DOI: 10.1186/s40644-024-00769-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 08/31/2024] [Indexed: 10/09/2024] Open
Abstract
Gliomas and Glioblastomas represent a significant portion of central nervous system (CNS) tumors associated with high mortality rates and variable prognosis. In 2021, the World Health Organization (WHO) updated its Glioma classification criteria, most notably incorporating molecular markers including CDKN2A/B homozygous deletion, TERT promoter mutation, EGFR amplification, + 7/-10 chromosome copy number changes, and others into the grading and classification of adult and pediatric Gliomas. The inclusion of these markers and the corresponding introduction of new Glioma subtypes has allowed for more specific tailoring of clinical interventions and has inspired a new wave of Radiogenomic studies seeking to leverage medical imaging information to explore the diagnostic and prognostic implications of these new biomarkers. Radiomics, deep learning, and combined approaches have enabled the development of powerful computational tools for MRI analysis correlating imaging characteristics with various molecular biomarkers integrated into the updated WHO CNS-5 guidelines. Recent studies have leveraged these methods to accurately classify Gliomas in accordance with these updated molecular-based criteria based solely on non-invasive MRI, demonstrating the great promise of Radiogenomic tools. In this review, we explore the relative benefits and drawbacks of these computational frameworks and highlight the technical and clinical innovations presented by recent studies in the landscape of fast evolving molecular-based Glioma subtyping. Furthermore, the potential benefits and challenges of incorporating these tools into routine radiological workflows, aiming to enhance patient care and optimize clinical outcomes in the evolving field of CNS tumor management, have been highlighted.
Collapse
Affiliation(s)
- Gagandeep Singh
- Neuroradiology Division, Columbia University Irving Medical Center, New York, NY, USA.
| | - Annie Singh
- Atal Bihari Vajpayee Institute of Medical Sciences, New Delhi, India
| | - Joseph Bae
- Department of Biomedical Informatics, Stony Brook University, Stony Brook, USA
| | - Sunil Manjila
- Department of Neurological Surgery, Garden City Hospital, Garden City, MI, USA
| | - Vadim Spektor
- Neuroradiology Division, Columbia University Irving Medical Center, New York, NY, USA
| | - Prateek Prasanna
- Department of Biomedical Informatics, Stony Brook University, Stony Brook, USA
| | - Angela Lignelli
- Neuroradiology Division, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
33
|
Zhang Y, Huang W, Jiao H, Kang L. PET radiomics in lung cancer: advances and translational challenges. EJNMMI Phys 2024; 11:81. [PMID: 39361110 PMCID: PMC11450131 DOI: 10.1186/s40658-024-00685-5] [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: 11/19/2023] [Accepted: 09/26/2024] [Indexed: 10/06/2024] Open
Abstract
Radiomics is an emerging field of medical imaging that aims at improving the accuracy of diagnosis, prognosis, treatment planning and monitoring non-invasively through the automated or semi-automated quantitative analysis of high-dimensional image features. Specifically in the field of nuclear medicine, radiomics utilizes imaging methods such as positron emission tomography (PET) and single photon emission computed tomography (SPECT) to evaluate biomarkers related to metabolism, blood flow, cellular activity and some biological pathways. Lung cancer ranks among the leading causes of cancer-related deaths globally, and radiomics analysis has shown great potential in guiding individualized therapy, assessing treatment response, and predicting clinical outcomes. In this review, we summarize the current state-of-the-art radiomics progress in lung cancer, highlighting the potential benefits and existing limitations of this approach. The radiomics workflow was introduced first including image acquisition, segmentation, feature extraction, and model building. Then the published literatures were described about radiomics-based prediction models for lung cancer diagnosis, differentiation, prognosis and efficacy evaluation. Finally, we discuss current challenges and provide insights into future directions and potential opportunities for integrating radiomics into routine clinical practice.
Collapse
Affiliation(s)
- Yongbai Zhang
- Department of Nuclear Medicine, Peking University First Hospital, No. 8 Xishiku Str., Xicheng Dist, Beijing, 100034, China
| | - Wenpeng Huang
- Department of Nuclear Medicine, Peking University First Hospital, No. 8 Xishiku Str., Xicheng Dist, Beijing, 100034, China
| | - Hao Jiao
- Department of Nuclear Medicine, Peking University First Hospital, No. 8 Xishiku Str., Xicheng Dist, Beijing, 100034, China
| | - Lei Kang
- Department of Nuclear Medicine, Peking University First Hospital, No. 8 Xishiku Str., Xicheng Dist, Beijing, 100034, China.
| |
Collapse
|
34
|
Sanchez I, Rahman R. Radiogenomics as an Integrated Approach to Glioblastoma Precision Medicine. Curr Oncol Rep 2024; 26:1213-1222. [PMID: 39009914 PMCID: PMC11480134 DOI: 10.1007/s11912-024-01580-z] [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] [Accepted: 07/05/2024] [Indexed: 07/17/2024]
Abstract
PURPOSE OF REVIEW Isocitrate dehydrogenase wild-type glioblastoma is the most aggressive primary brain tumour in adults. Its infiltrative nature and heterogeneity confer a dismal prognosis, despite multimodal treatment. Precision medicine is increasingly advocated to improve survival rates in glioblastoma management; however, conventional neuroimaging techniques are insufficient in providing the detail required for accurate diagnosis of this complex condition. RECENT FINDINGS Advanced magnetic resonance imaging allows more comprehensive understanding of the tumour microenvironment. Combining diffusion and perfusion magnetic resonance imaging to create a multiparametric scan enhances diagnostic power and can overcome the unreliability of tumour characterisation by standard imaging. Recent progress in deep learning algorithms establishes their remarkable ability in image-recognition tasks. Integrating these with multiparametric scans could transform the diagnosis and monitoring of patients by ensuring that the entire tumour is captured. As a corollary, radiomics has emerged as a powerful approach to offer insights into diagnosis, prognosis, treatment, and tumour response through extraction of information from radiological scans, and transformation of these tumour characteristics into quantitative data. Radiogenomics, which links imaging features with genomic profiles, has exhibited its ability in characterising glioblastoma, and determining therapeutic response, with the potential to revolutionise management of glioblastoma. The integration of deep learning algorithms into radiogenomic models has established an automated, highly reproducible means to predict glioblastoma molecular signatures, further aiding prognosis and targeted therapy. However, challenges including lack of large cohorts, absence of standardised guidelines and the 'black-box' nature of deep learning algorithms, must first be overcome before this workflow can be applied in clinical practice.
Collapse
Affiliation(s)
- Isabella Sanchez
- Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Ruman Rahman
- Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham, NG7 2RD, UK.
| |
Collapse
|
35
|
Le Guillou Horn XM, Lecellier F, Giraud C, Naudin M, Fayolle P, Thomarat C, Fernandez-Maloigne C, Guillevin R. From Voxel to Gene: A Scoping Review on MRI Radiogenomics' Artificial Intelligence Predictions in Adult Gliomas and Glioblastomas-The Promise of Virtual Biopsy? Biomedicines 2024; 12:2156. [PMID: 39335670 PMCID: PMC11429468 DOI: 10.3390/biomedicines12092156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/12/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Gliomas, including the most severe form known as glioblastomas, are primary brain tumors arising from glial cells, with significant impact on adults, particularly men aged 45 to 70. Recent advancements in the WHO (World Health Organization) classification now correlate genetic markers with glioma phenotypes, enhancing diagnostic precision and therapeutic strategies. AIMS AND METHODS This scoping review aims to evaluate the current state of deep learning (DL) applications in the genetic characterization of adult gliomas, addressing the potential of these technologies for a reliable virtual biopsy. RESULTS We reviewed 17 studies, analyzing the evolution of DL algorithms from fully convolutional networks to more advanced architectures (ResNet and DenseNet). The methods involved various validation techniques, including k-fold cross-validation and external dataset validation. CONCLUSIONS Our findings highlight significant variability in reported performance, largely due to small, homogeneous datasets and inconsistent validation methods. Despite promising results, particularly in predicting individual genetic traits, the lack of robust external validation limits the generalizability of these models. Future efforts should focus on developing larger, more diverse datasets and integrating multidisciplinary collaboration to enhance model reliability. This review underscores the potential of DL in advancing glioma characterization, paving the way for more precise, non-invasive diagnostic tools. The development of a robust algorithm capable of predicting the somatic genetics of gliomas or glioblastomas could accelerate the diagnostic process and inform therapeutic decisions more quickly, while maintaining the same level of accuracy as the traditional diagnostic pathway, which involves invasive tumor biopsies.
Collapse
Affiliation(s)
- Xavier Maximin Le Guillou Horn
- Laboratoire de Mathématique Appliquées LMA, Labcom i3M, Université de Poitiers, CNRS UMR 7348, F-86000 Poitiers, France (P.F.)
- Service de Génétique Médicale, CHU de Poitiers, F-86000 Poitiers, France
| | - François Lecellier
- Laboratoire XLIM, Université de Poitiers, CNRS UMR 7252, F-86000 Poitiers, France
| | - Clement Giraud
- Laboratoire de Mathématique Appliquées LMA, Labcom i3M, Université de Poitiers, CNRS UMR 7348, F-86000 Poitiers, France (P.F.)
- Plateforme Ultra-Haut Champ 3T-7T, Laboratoire de Mathématique Appliquées LMA, Labcom i3M, CHU de Poitiers, Université de Poitiers, CNRS UMR 7348, F-86000 Poitiers, France (R.G.)
| | - Mathieu Naudin
- Plateforme Ultra-Haut Champ 3T-7T, Laboratoire de Mathématique Appliquées LMA, Labcom i3M, CHU de Poitiers, Université de Poitiers, CNRS UMR 7348, F-86000 Poitiers, France (R.G.)
| | - Pierre Fayolle
- Laboratoire de Mathématique Appliquées LMA, Labcom i3M, Université de Poitiers, CNRS UMR 7348, F-86000 Poitiers, France (P.F.)
- Plateforme Ultra-Haut Champ 3T-7T, Laboratoire de Mathématique Appliquées LMA, Labcom i3M, CHU de Poitiers, Université de Poitiers, CNRS UMR 7348, F-86000 Poitiers, France (R.G.)
| | - Céline Thomarat
- Plateforme Ultra-Haut Champ 3T-7T, Laboratoire de Mathématique Appliquées LMA, Labcom i3M, CHU de Poitiers, Université de Poitiers, CNRS UMR 7348, F-86000 Poitiers, France (R.G.)
| | | | - Rémy Guillevin
- Plateforme Ultra-Haut Champ 3T-7T, Laboratoire de Mathématique Appliquées LMA, Labcom i3M, CHU de Poitiers, Université de Poitiers, CNRS UMR 7348, F-86000 Poitiers, France (R.G.)
- Service de Radiologie, CHU de Poitiers, F-86000 Poitiers, France
| |
Collapse
|
36
|
Zheng B, Zhao Z, Zheng P, Liu Q, Li S, Jiang X, Huang X, Ye Y, Wang H. The current state of MRI-based radiomics in pituitary adenoma: promising but challenging. Front Endocrinol (Lausanne) 2024; 15:1426781. [PMID: 39371931 PMCID: PMC11449739 DOI: 10.3389/fendo.2024.1426781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 08/30/2024] [Indexed: 10/08/2024] Open
Abstract
In the clinical diagnosis and treatment of pituitary adenomas, MRI plays a crucial role. However, traditional manual interpretations are plagued by inter-observer variability and limitations in recognizing details. Radiomics, based on MRI, facilitates quantitative analysis by extracting high-throughput data from images. This approach elucidates correlations between imaging features and pituitary tumor characteristics, thereby establishing imaging biomarkers. Recent studies have demonstrated the extensive application of radiomics in differential diagnosis, subtype identification, consistency evaluation, invasiveness assessment, and treatment response in pituitary adenomas. This review succinctly presents the general workflow of radiomics, reviews pertinent literature with a summary table, and provides a comparative analysis with traditional methods. We further elucidate the connections between radiological features and biological findings in the field of pituitary adenoma. While promising, the clinical application of radiomics still has a considerable distance to traverse, considering the issues with reproducibility of imaging features and the significant heterogeneity in pituitary adenoma patients.
Collapse
Affiliation(s)
- Baoping Zheng
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhen Zhao
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pingping Zheng
- Department of Neurosurgery, People’s Hospital of Biyang County, Zhumadian, China
| | - Qiang Liu
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuang Li
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaobing Jiang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xing Huang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Youfan Ye
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haijun Wang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
37
|
Huang YR, Fan HQ, Kuang YY, Wang P, Lu S. The Relationship Between the Molecular Phenotypes of Brain Gliomas and the Imaging Features and Sensitivity of Radiotherapy and Chemotherapy. Clin Oncol (R Coll Radiol) 2024; 36:541-551. [PMID: 38821723 DOI: 10.1016/j.clon.2024.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 02/28/2024] [Accepted: 05/10/2024] [Indexed: 06/02/2024]
Abstract
Gliomas are the most common primary malignant tumors of the brain, accounting for about 80% of all central nervous system malignancies. With the development of molecular biology, the molecular phenotypes of gliomas have been shown to be closely related to the process of diagnosis and treatment. The molecular phenotype of glioma also plays an important role in guiding treatment plans and evaluating treatment effects and prognosis. However, due to the heterogeneity of the tumors and the trauma associated with the surgical removal of tumor tissue, the application of molecular phenotyping in glioma is limited. With the development of imaging technology, functional magnetic resonance imaging (MRI) can provide structural and function information about tumors in a noninvasive and radiation-free manner. MRI is very important for the diagnosis of intracranial lesions. In recent years, with the development of the technology for tumor molecular diagnosis and imaging, the use of molecular phenotype information and imaging procedures to evaluate the treatment outcome of tumors has become a hot topic. By reviewing the related literature on glioma treatment and molecular typing that has been published in the past 20 years, and referring to the latest 2020 NCCN treatment guidelines, summarizing the imaging characteristic and sensitivity of radiotherapy and chemotherapy of different molecular phenotypes of glioma. In this article, we briefly review the imaging characteristics of different molecular phenotypes in gliomas and their relationship with radiosensitivity and chemosensitivity of gliomas.
Collapse
Affiliation(s)
- Y-R Huang
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - H-Q Fan
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Y-Y Kuang
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - P Wang
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - S Lu
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China.
| |
Collapse
|
38
|
Usuzaki T, Inamori R, Shizukuishi T, Morishita Y, Takagi H, Ishikuro M, Obara T, Takase K. Predicting isocitrate dehydrogenase status among adult patients with diffuse glioma using patient characteristics, radiomic features, and magnetic resonance imaging: Multi-modal analysis by variable vision transformer. Magn Reson Imaging 2024; 111:266-276. [PMID: 38815636 DOI: 10.1016/j.mri.2024.05.012] [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: 04/21/2024] [Revised: 05/14/2024] [Accepted: 05/22/2024] [Indexed: 06/01/2024]
Abstract
OBJECTIVES To evaluate the performance of the multimodal model, termed variable Vision Transformer (vViT), in the task of predicting isocitrate dehydrogenase (IDH) status among adult patients with diffuse glioma. MATERIALS AND METHODS vViT was designed to predict IDH status using patient characteristics (sex and age), radiomic features, and contrast-enhanced T1-weighted images (CE-T1WI). Radiomic features were extracted from each enhancing tumor (ET), necrotic tumor core (NCR), and peritumoral edematous/infiltrated tissue (ED). CE-T1WI were split into four images and input to vViT. In the training, internal test, and external test, 271 patients with 1070 images (535 IDH wildtype, 535 IDH mutant), 35 patients with 194 images (97 IDH wildtype, 97 IDH mutant), and 291 patients with 872 images (436 IDH wildtype, 436 IDH mutant) were analyzed, respectively. Metrics including accuracy and AUC-ROC were calculated for the internal and external test datasets. Permutation importance analysis combined with the Mann-Whitney U test was performed to compare inputs. RESULTS For the internal test dataset, vViT correctly predicted IDH status for all patients. For the external test dataset, an accuracy of 0.935 (95% confidence interval; 0.913-0.945) and AUC-ROC of 0.887 (0.798-0.956) were obtained. For both internal and external test datasets, CE-T1WI ET radiomic features and patient characteristics had higher importance than other inputs (p < 0.05). CONCLUSIONS The vViT has the potential to be a competent model in predicting IDH status among adult patients with diffuse glioma. Our results indicate that age, sex, and CE-T1WI ET radiomic features have key information in estimating IDH status.
Collapse
Affiliation(s)
- Takuma Usuzaki
- Department of Diagnostic Radiology, Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8574, Japan; Miyagi Cancer Center, Miyagi, Japan
| | - Ryusei Inamori
- Radiological Imaging and Informatics, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takashi Shizukuishi
- Department of Diagnostic Radiology, Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8574, Japan
| | - Yohei Morishita
- Department of Diagnostic Radiology, Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8574, Japan
| | - Hidenobu Takagi
- Department of Diagnostic Radiology, Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8574, Japan; Department of Advanced MRI Collaborative Research, Graduate School of Medicine, Sendai, Japan
| | - Mami Ishikuro
- Tohoku University Graduate School of Medicine, Division of Molecular Epidemiology, Sendai, Japan
| | - Taku Obara
- Tohoku University Graduate School of Medicine, Division of Molecular Epidemiology, Sendai, Japan; Tohoku University Graduate School of Medicine, Division of Molecular Epidemiology, Department of Preventive Medicine and Epidemiology, Tohoku Medical Megabank Organization, Sendai, Japan; Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Japan
| | - Kei Takase
- Department of Diagnostic Radiology, Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8574, Japan
| |
Collapse
|
39
|
Kudus K, Wagner MW, Namdar K, Bennett J, Nobre L, Tabori U, Hawkins C, Ertl-Wagner BB, Khalvati F. Beyond hand-crafted features for pretherapeutic molecular status identification of pediatric low-grade gliomas. Sci Rep 2024; 14:19102. [PMID: 39154039 PMCID: PMC11330469 DOI: 10.1038/s41598-024-69870-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 08/09/2024] [Indexed: 08/19/2024] Open
Abstract
The use of targeted agents in the treatment of pediatric low-grade gliomas (pLGGs) relies on the determination of molecular status. It has been shown that genetic alterations in pLGG can be identified non-invasively using MRI-based radiomic features or convolutional neural networks (CNNs). We aimed to build and assess a combined radiomics and CNN non-invasive pLGG molecular status identification model. This retrospective study used the tumor regions, manually segmented from T2-FLAIR MR images, of 336 patients treated for pLGG between 1999 and 2018. We designed a CNN and Random Forest radiomics model, along with a model relying on a combination of CNN and radiomic features, to predict the genetic status of pLGG. Additionally, we investigated whether CNNs could predict radiomic feature values from MR images. The combined model (mean AUC: 0.824) outperformed the radiomics model (0.802) and CNN (0.764). The differences in model performance were statistically significant (p-values < 0.05). The CNN was able to learn predictive radiomic features such as surface-to-volume ratio (average correlation: 0.864), and difference matrix dependence non-uniformity normalized (0.924) well but was unable to learn others such as run-length matrix variance (- 0.017) and non-uniformity normalized (- 0.042). Our results show that a model relying on both CNN and radiomic-based features performs better than either approach separately in differentiating the genetic status of pLGGs, and that CNNs are unable to express all handcrafted features.
Collapse
Affiliation(s)
- Kareem Kudus
- Neurosciences & Mental Health Research Program, The Hospital for Sick Children, Toronto, Canada
- Institute of Medical Science, University of Toronto, Toronto, Canada
| | - Matthias W Wagner
- Department of Diagnostic & Interventional Radiology, The Hospital for Sick Children, Toronto, Canada
- Department of Diagnostic and Interventional Neuroradiology, University Hospital Augsburg, Augsburg, Germany
| | - Khashayar Namdar
- Neurosciences & Mental Health Research Program, The Hospital for Sick Children, Toronto, Canada
- Institute of Medical Science, University of Toronto, Toronto, Canada
| | - Julie Bennett
- Division of Hematology and Oncology, The Hospital for Sick Children, Toronto, Canada
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Canada
- Department of Pediatrics, University of Toronto, Toronto, Canada
| | - Liana Nobre
- Department of Paediatrics, University of Alberta, Edmonton, Canada
- Division of Immunology, Hematology/Oncology and Palliative Care, Stollery Children's Hospital, Edmonton, Canada
| | - Uri Tabori
- Division of Hematology and Oncology, The Hospital for Sick Children, Toronto, Canada
| | - Cynthia Hawkins
- Paediatric Laboratory Medicine, Division of Pathology, The Hospital for Sick Children, Toronto, Canada
| | - Birgit Betina Ertl-Wagner
- Neurosciences & Mental Health Research Program, The Hospital for Sick Children, Toronto, Canada
- Institute of Medical Science, University of Toronto, Toronto, Canada
- Department of Diagnostic & Interventional Radiology, The Hospital for Sick Children, Toronto, Canada
- Department of Medical Imaging, University of Toronto, Toronto, Canada
| | - Farzad Khalvati
- Neurosciences & Mental Health Research Program, The Hospital for Sick Children, Toronto, Canada.
- Institute of Medical Science, University of Toronto, Toronto, Canada.
- Department of Diagnostic & Interventional Radiology, The Hospital for Sick Children, Toronto, Canada.
- Department of Medical Imaging, University of Toronto, Toronto, Canada.
- Department of Computer Science, University of Toronto, Toronto, Canada.
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Canada.
| |
Collapse
|
40
|
Wu X, Zhang S, Zhang Z, He Z, Xu Z, Wang W, Jin Z, You J, Guo Y, Zhang L, Huang W, Wang F, Liu X, Yan D, Cheng J, Yan J, Zhang S, Zhang B. Biologically interpretable multi-task deep learning pipeline predicts molecular alterations, grade, and prognosis in glioma patients. NPJ Precis Oncol 2024; 8:181. [PMID: 39152182 PMCID: PMC11329669 DOI: 10.1038/s41698-024-00670-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 08/01/2024] [Indexed: 08/19/2024] Open
Abstract
Deep learning models have been developed for various predictions in glioma; yet, they were constrained by manual segmentation, task-specific design, or a lack of biological interpretation. Herein, we aimed to develop an end-to-end multi-task deep learning (MDL) pipeline that can simultaneously predict molecular alterations and histological grade (auxiliary tasks), as well as prognosis (primary task) in gliomas. Further, we aimed to provide the biological mechanisms underlying the model's predictions. We collected multiscale data including baseline MRI images from 2776 glioma patients across two private (FAHZU and HPPH, n = 1931) and three public datasets (TCGA, n = 213; UCSF, n = 410; and EGD, n = 222). We trained and internally validated the MDL model using our private datasets, and externally validated it using the three public datasets. We used the model-predicted deep prognosis score (DPS) to stratify patients into low-DPS and high-DPS subtypes. Additionally, a radio-multiomics analysis was conducted to elucidate the biological basis of the DPS. In the external validation cohorts, the MDL model achieved average areas under the curve of 0.892-0.903, 0.710-0.894, and 0.850-0.879 for predicting IDH mutation status, 1p/19q co-deletion status, and tumor grade, respectively. Moreover, the MDL model yielded a C-index of 0.723 in the TCGA and 0.671 in the UCSF for the prediction of overall survival. The DPS exhibits significant correlations with activated oncogenic pathways, immune infiltration patterns, specific protein expression, DNA methylation, tumor mutation burden, and tumor-stroma ratio. Accordingly, our work presents an accurate and biologically meaningful tool for predicting molecular subtypes, tumor grade, and survival outcomes in gliomas, which provides personalized clinical decision-making in a global and non-invasive manner.
Collapse
Affiliation(s)
- Xuewei Wu
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Shuaitong Zhang
- School of Medical Technology, Beijing Institute of Technology, Beijing, China
| | - Zhenyu Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zicong He
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Zexin Xu
- School of Medical Technology, Beijing Institute of Technology, Beijing, China
| | - Weiwei Wang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhe Jin
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Jingjing You
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Yang Guo
- Department of Neurosurgery, The Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Lu Zhang
- 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
| | - Fei Wang
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Xianzhi Liu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Dongming Yan
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jingliang Cheng
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jing Yan
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 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
|
41
|
Habibi MA, Dinpazhouh A, Aliasgary A, Mirjani MS, Mousavinasab M, Ahmadi MR, Minaee P, Eazi S, Shafizadeh M, Gurses ME, Lu VM, Berke CN, Ivan ME, Komotar RJ, Shah AH. Predicting telomerase reverse transcriptase promoter mutation in glioma: A systematic review and diagnostic meta-analysis on machine learning algorithms. Neuroradiol J 2024:19714009241269526. [PMID: 39103206 PMCID: PMC11571522 DOI: 10.1177/19714009241269526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 07/01/2024] [Accepted: 07/06/2024] [Indexed: 08/07/2024] Open
Abstract
BACKGROUND Glioma is one of the most common primary brain tumors. The presence of the telomerase reverse transcriptase promoter (pTERT) mutation is associated with a better prognosis. This study aims to investigate the TERT mutation in patients with glioma using machine learning (ML) algorithms on radiographic imaging. METHOD This study was prepared according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. The electronic databases of PubMed, Embase, Scopus, and Web of Science were searched from inception to August 1, 2023. The statistical analysis was performed using the MIDAS package of STATA v.17. RESULTS A total of 22 studies involving 5371 patients were included for data extraction, with data synthesis based on 11 reports. The analysis revealed a pooled sensitivity of 0.86 (95% CI: 0.78-0.92) and a specificity of 0.80 (95% CI 0.72-0.86). The positive and negative likelihood ratios were 4.23 (95% CI: 2.99-5.99) and 0.18 (95% CI: 0.11-0.29), respectively. The pooled diagnostic score was 3.18 (95% CI: 2.45-3.91), with a diagnostic odds ratio 24.08 (95% CI: 11.63-49.87). The Summary Receiver Operating Characteristic (SROC) curve had an area under the curve (AUC) of 0.89 (95% CI: 0.86-0.91). CONCLUSION The study suggests that ML can predict TERT mutation status in glioma patients. ML models showed high sensitivity (0.86) and moderate specificity (0.80), aiding disease prognosis and treatment planning. However, further development and improvement of ML models are necessary for better performance metrics and increased reliability in clinical practice.
Collapse
Affiliation(s)
- Mohammad Amin Habibi
- Department of Neurosurgery, Shariati Hospital, Tehran University of Medical Science, Tehran, Iran
| | - Ali Dinpazhouh
- Student Research Committee, Faculty of Medicine, Qom University of Medical Science, Qom, Iran
| | - Aliakbar Aliasgary
- Student Research Committee, Faculty of Medicine, Qom University of Medical Science, Qom, Iran
| | - Mohammad Sina Mirjani
- Student Research Committee, Faculty of Medicine, Qom University of Medical Science, Qom, Iran
| | - Mehdi Mousavinasab
- Student Research Committee, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Mohammad Reza Ahmadi
- Student Research Committee, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Poriya Minaee
- Student Research Committee, Faculty of Medicine, Qom University of Medical Science, Qom, Iran
| | - SeyedMohammad Eazi
- Student Research Committee, Faculty of Medicine, Qom University of Medical Science, Qom, Iran
| | - Milad Shafizadeh
- Department of Neurosurgery, Shariati Hospital, Tehran University of Medical Science, Tehran, Iran
| | - Muhammet Enes Gurses
- Department of Neurosurgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Victor M. Lu
- Department of Neurosurgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Chandler N. Berke
- Department of Neurosurgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Michael E. Ivan
- Department of Neurosurgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Ricardo J. Komotar
- Department of Neurosurgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Ashish H. Shah
- Department of Neurosurgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| |
Collapse
|
42
|
Fan L, Wu H, Wu Y, Wu S, Zhao J, Zhu X. Preoperative prediction of rectal Cancer staging combining MRI deep transfer learning, radiomics features, and clinical factors: accurate differentiation from stage T2 to T3. BMC Gastroenterol 2024; 24:247. [PMID: 39103772 DOI: 10.1186/s12876-024-03316-6] [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: 03/31/2024] [Accepted: 07/04/2024] [Indexed: 08/07/2024] Open
Abstract
BACKGROUND This study evaluates the efficacy of integrating MRI deep transfer learning, radiomic signatures, and clinical variables to accurately preoperatively differentiate between stage T2 and T3 rectal cancer. METHODS We included 361 patients with pathologically confirmed stage T2 or T3 rectal cancer, divided into a training set (252 patients) and a test set (109 patients) at a 7:3 ratio. The study utilized features derived from deep transfer learning and radiomics, with Spearman rank correlation and the Least Absolute Shrinkage and Selection Operator (LASSO) regression techniques to reduce feature redundancy. Predictive models were developed using Logistic Regression (LR), Random Forest (RF), Decision Tree (DT), and Support Vector Machine (SVM), selecting the best-performing model for a comprehensive predictive framework incorporating clinical data. RESULTS After removing redundant features, 24 key features were identified. In the training set, the area under the curve (AUC)values for LR, RF, DT, and SVM were 0.867, 0.834, 0.900, and 0.944, respectively; in the test set, they were 0.847, 0.803, 0.842, and 0.910, respectively. The combined model, using SVM and clinical variables, achieved AUCs of 0.946 in the trainingset and 0.920 in the test set. CONCLUSION The study confirms the utility of a combined model of MRI deep transfer learning, radiomic features, and clinical factors for preoperative classification of stage T2 vs. T3 rectal cancer, offering significant technological support for precise diagnosis and potential clinical application.
Collapse
Affiliation(s)
- Lifang Fan
- School of Medical Imageology, Wannan Medical College, Wuhu, 241002, Anhui, China
- Anhui Province Key Laboratory of Cancer Translational Medicine, Bengbu Medical University, 2600 Donghai Avenue, Bengbu, Anhui, 233030, China
| | - Huazhang Wu
- Anhui Province Key Laboratory of Cancer Translational Medicine, Bengbu Medical University, 2600 Donghai Avenue, Bengbu, Anhui, 233030, China
| | - Yimin Wu
- Department of Ultrasound, The Second People's Hospital, WuHu Hospital, East China Normal University, Wuhu, Anhui, 241001, China
| | - Shujian Wu
- Department of Radiology, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, China
| | - Jinsong Zhao
- School of Medical Imageology, Wannan Medical College, Wuhu, 241002, Anhui, China.
| | - Xiangming Zhu
- Department of Ultrasound, Yijishan Hospital of Wannan Medical College, No.2 Zheshan West Road, Jinghu District, Wuhu, Anhui Province, 241001, China.
| |
Collapse
|
43
|
Yang X, Niu W, Wu K, Li X, Hou H, Tan Y, Wang X, Yang G, Wang L, Zhang H. Diffusion kurtosis imaging-based habitat analysis identifies high-risk molecular subtypes and heterogeneity matching in diffuse gliomas. Ann Clin Transl Neurol 2024; 11:2073-2087. [PMID: 38887966 PMCID: PMC11330218 DOI: 10.1002/acn3.52128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 05/14/2024] [Accepted: 06/02/2024] [Indexed: 06/20/2024] Open
Abstract
OBJECTIVE High-risk types of diffuse gliomas in adults include isocitrate dehydrogenase (IDH) wild-type glioblastomas and grade 4 astrocytomas. Achieving noninvasive prediction of high-risk molecular subtypes of gliomas is important for personalized and precise diagnosis and treatment. METHODS We retrospectively collected data from 116 patients diagnosed with adult diffuse gliomas. Multiple high-risk molecular markers were tested, and various habitat models and whole-tumor models were constructed based on preoperative routine and diffusion kurtosis imaging (DKI) sequences to predict high-risk molecular subtypes of gliomas. Feature selection and model construction utilized Least absolute shrinkage and selection operator (LASSO) and support vector machine (SVM). Finally, the Wilcoxon rank-sum test was employed to explore the correlation between habitat quantitative features (intra-tumor heterogeneity score,ITH score) and heterogeneity, as well as high-risk molecular subtypes. RESULTS The results showed that the habitat analysis model based on DKI performed remarkably well (with AUC values reaching 0.977 and 0.902 in the training and test sets, respectively). The model's performance was further enhanced when combined with clinical variables. (The AUC values were 0.994 and 0.920, respectively.) Additionally, we found a close correlation between ITH score and heterogeneity, with statistically significant differences observed between high-risk and non-high-risk molecular subtypes. INTERPRETATION The habitat model based on DKI is an ideal means for preoperatively predicting high-risk molecular subtypes of gliomas, holding significant value for noninvasively alerting malignant gliomas and those with malignant transformation potential.
Collapse
Affiliation(s)
- Xiangli Yang
- Department of RadiologyFirst Hospital of Shanxi Medical UniversityTaiyuan030001China
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi HospitalTaiyuan030032China
- College of Medical Imaging, Shanxi Medical UniversityTaiyuan030001China
| | - Wenju Niu
- College of Medical Imaging, Shanxi Medical UniversityTaiyuan030001China
| | - Kai Wu
- Department of Information ManagementFirst Hospital of Shanxi Medical UniversityTaiyuan030001China
| | - Xiang Li
- College of Medical Imaging, Shanxi Medical UniversityTaiyuan030001China
| | - Heng Hou
- Department of RadiologyFirst Hospital of Shanxi Medical UniversityTaiyuan030001China
| | - Yan Tan
- Department of RadiologyFirst Hospital of Shanxi Medical UniversityTaiyuan030001China
| | - Xiaochun Wang
- Department of RadiologyFirst Hospital of Shanxi Medical UniversityTaiyuan030001China
| | - Guoqiang Yang
- Department of RadiologyFirst Hospital of Shanxi Medical UniversityTaiyuan030001China
- College of Medical Imaging, Shanxi Medical UniversityTaiyuan030001China
- Shanxi Key Laboratory of Intelligent Imaging and NanomedicineFirst Hospital of Shanxi Medical UniversityTaiyuan030001China
| | - Lei Wang
- Beijing Tiantan HospitalCapital Medical UniversityBeijing100050China
| | - Hui Zhang
- Department of RadiologyFirst Hospital of Shanxi Medical UniversityTaiyuan030001China
- College of Medical Imaging, Shanxi Medical UniversityTaiyuan030001China
- Shanxi Key Laboratory of Intelligent Imaging and NanomedicineFirst Hospital of Shanxi Medical UniversityTaiyuan030001China
- Intelligent Imaging Big Data and Functional Nano‐imaging Engineering Research Center of Shanxi ProvinceFirst Hospital of Shanxi Medical UniversityTaiyuan030001China
| |
Collapse
|
44
|
Rudà R, Horbinski C, van den Bent M, Preusser M, Soffietti R. IDH inhibition in gliomas: from preclinical models to clinical trials. Nat Rev Neurol 2024; 20:395-407. [PMID: 38760442 DOI: 10.1038/s41582-024-00967-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2024] [Indexed: 05/19/2024]
Abstract
Gliomas are the most common malignant primary brain tumours in adults and cannot usually be cured with standard cancer treatments. Gliomas show intratumoural and intertumoural heterogeneity at the histological and molecular levels, and they frequently contain mutations in the isocitrate dehydrogenase 1 (IDH1) or IDH2 gene. IDH-mutant adult-type diffuse gliomas are subdivided into grade 2, 3 or 4 IDH-mutant astrocytomas and grade 2 or 3 IDH-mutant, 1p19q-codeleted oligodendrogliomas. The product of the mutated IDH genes, D-2-hydroxyglutarate (D-2-HG), induces global DNA hypermethylation and interferes with immunity, leading to stimulation of tumour growth. Selective inhibitors of mutant IDH, such as ivosidenib and vorasidenib, have been shown to reduce D-2-HG levels and induce cellular differentiation in preclinical models and to induce MRI-detectable responses in early clinical trials. The phase III INDIGO trial has demonstrated superiority of vorasidenib, a brain-penetrant pan-mutant IDH inhibitor, over placebo in people with non-enhancing grade 2 IDH-mutant gliomas following surgery. In this Review, we describe the pathway of development of IDH inhibitors in IDH-mutant low-grade gliomas from preclinical models to clinical trials. We discuss the practice-changing implications of the INDIGO trial and consider new avenues of investigation in the field of IDH-mutant gliomas.
Collapse
Affiliation(s)
- Roberta Rudà
- Division of Neuro-Oncology, Department of Neuroscience 'Rita Levi Montalcini', University of Turin, Turin, Italy.
| | - Craig Horbinski
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Martin van den Bent
- Brain Tumour Center at Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Matthias Preusser
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Riccardo Soffietti
- Division of Neuro-Oncology, Department of Neuroscience 'Rita Levi Montalcini', University of Turin, Turin, Italy
| |
Collapse
|
45
|
Zeng Y, Liu N, Yang X, Huang C, Liu M. Enhanced Multimodal Brain Tumor Classification in MR Images using 2D ResNet as backbone with Explicit Tumor Size Information. J Cancer 2024; 15:4275-4286. [PMID: 38947386 PMCID: PMC11212098 DOI: 10.7150/jca.95987] [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: 03/06/2024] [Accepted: 05/02/2024] [Indexed: 07/02/2024] Open
Abstract
It's a major public health problem of global concern that malignant gliomas tend to grow rapidly and infiltrate surrounding tissues. Accurate grading of the tumor can determine the degree of malignancy to formulate the best treatment plan, which can eliminate the tumor or limit widespread metastasis of the tumor, saving the patient's life and improving their prognosis. To more accurately predict the grading of gliomas, we proposed a novel method of combining the advantages of 2D and 3D Convolutional Neural Networks for tumor grading by multimodality on Magnetic Resonance Imaging. The core of the innovation lies in our combination of tumor 3D information extracted from multimodal data with those obtained from a 2D ResNet50 architecture. It solves both the lack of temporal-spatial information provided by 3D imaging in 2D convolutional neural networks and avoids more noise from too much information in 3D convolutional neural networks, which causes serious overfitting problems. Incorporating explicit tumor 3D information, such as tumor volume and surface area, enhances the grading model's performance and addresses the limitations of both approaches. By fusing information from multiple modalities, the model achieves a more precise and accurate characterization of tumors. The model I s trained and evaluated using two publicly available brain glioma datasets, achieving an AUC of 0.9684 on the validation set. The model's interpretability is enhanced through heatmaps, which highlight the tumor region. The proposed method holds promise for clinical application in tumor grading and contributes to the field of medical diagnostics for prediction.
Collapse
Affiliation(s)
- Yunhao Zeng
- School of Information and Software Engineering, University of Electronic Science and Technology of China, Chengdu 611731, China
- Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Quzhou 324000, China
| | - Nianbo Liu
- Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Quzhou 324000, China
- Quzhou People's Hospital, Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou 324000, China
| | - Xinduoji Yang
- School of Information and Software Engineering, University of Electronic Science and Technology of China, Chengdu 611731, China
- Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Quzhou 324000, China
| | - Chenke Huang
- School of Information and Software Engineering, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Ming Liu
- Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Quzhou 324000, China
- Quzhou People's Hospital, Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou 324000, China
| |
Collapse
|
46
|
Moon HH, Jeong J, Park JE, Kim N, Choi C, Kim Y, Song SW, Hong CK, Kim JH, Kim HS. Generative AI in glioma: Ensuring diversity in training image phenotypes to improve diagnostic performance for IDH mutation prediction. Neuro Oncol 2024; 26:1124-1135. [PMID: 38253989 PMCID: PMC11145451 DOI: 10.1093/neuonc/noae012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND This study evaluated whether generative artificial intelligence (AI)-based augmentation (GAA) can provide diverse and realistic imaging phenotypes and improve deep learning-based classification of isocitrate dehydrogenase (IDH) type in glioma compared with neuroradiologists. METHODS For model development, 565 patients (346 IDH-wildtype, 219 IDH-mutant) with paired contrast-enhanced T1 and FLAIR MRI scans were collected from tertiary hospitals and The Cancer Imaging Archive. Performance was tested on internal (119, 78 IDH-wildtype, 41 IDH-mutant [IDH1 and 2]) and external test sets (108, 72 IDH-wildtype, 36 IDH-mutant). GAA was developed using a score-based diffusion model and ResNet50 classifier. The optimal GAA was selected in comparison with the null model. Two neuroradiologists (R1, R2) assessed realism, diversity of imaging phenotypes, and predicted IDH mutation. The performance of a classifier trained with optimal GAA was compared with that of neuroradiologists using the area under the receiver operating characteristics curve (AUC). The effect of tumor size and contrast enhancement on GAA performance was tested. RESULTS Generated images demonstrated realism (Turing's test: 47.5-50.5%) and diversity indicating IDH type. Optimal GAA was achieved with augmentation with 110 000 generated slices (AUC: 0.938). The classifier trained with optimal GAA demonstrated significantly higher AUC values than neuroradiologists in both the internal (R1, P = .003; R2, P < .001) and external test sets (R1, P < .01; R2, P < .001). GAA with large-sized tumors or predominant enhancement showed comparable performance to optimal GAA (internal test: AUC 0.956 and 0.922; external test: 0.810 and 0.749). CONCLUSIONS The application of generative AI with realistic and diverse images provided better diagnostic performance than neuroradiologists for predicting IDH type in glioma.
Collapse
Affiliation(s)
- Hye Hyeon Moon
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Jiheon Jeong
- Department of Convergence Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
- Department of Biomedical Engineering, Asan Medical Institute of Convergence Science of Technology, Asan Medical Center, College of Medicine, University of Ulsan, Seoul, Korea
| | - Ji Eun Park
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Namkug Kim
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
- Department of Convergence Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Changyong Choi
- Department of Convergence Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
- Department of Biomedical Engineering, Asan Medical Institute of Convergence Science of Technology, Asan Medical Center, College of Medicine, University of Ulsan, Seoul, Korea
| | - Young‑Hoon Kim
- Department of Neurosurgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Sang Woo Song
- Department of Neurosurgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Chang-Ki Hong
- Department of Neurosurgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Jeong Hoon Kim
- Department of Neurosurgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Ho Sung Kim
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| |
Collapse
|
47
|
Wang S, Liu X, Jiang C, Kang W, Pan Y, Tang X, Luo Y, Gong J. CT-Based Super-Resolution Deep Learning Models with Attention Mechanisms for Predicting Spread Through Air Spaces of Solid or Part-Solid Lung Adenocarcinoma. Acad Radiol 2024; 31:2601-2609. [PMID: 38184418 DOI: 10.1016/j.acra.2023.12.034] [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: 12/20/2023] [Accepted: 12/20/2023] [Indexed: 01/08/2024]
Abstract
RATIONALE AND OBJECTIVES Spread through air space (STAS) is a novel invasive pattern of lung adenocarcinoma (LUAD), and preoperative knowledge of STAS status is helpful in choosing an appropriate surgical approach. MATERIALS AND METHODS This retrospective study collected and analyzed 602 patients diagnosed with LUAD from two medical centers: center 1 was randomly partitioned into training (n = 358) and validation cohorts (n = 154) at a 7:3 ratio; and center 2 was the external test cohort (n = 90). Super resolution was performed on all images to acquire high-resolution images, which were used to train the SE-ResNet50 model, before creating an equivalent parameter ResNet50 model. Disparities were compared between the two models using receiver operating characteristic curves, area under the curve, accuracy, precision, sensitivity, and specificity. RESULTS In this study, 512 and 90 patients with LUAD were enrolled from centers 1 and 2, respectively. The curve values of the SE-ResNet50 and ResNet50 models were compared for training, validation, and test cohorts, resulting in values of 0.933 vs 0.909, 0.783 vs 0.728, and 0.806 vs 0.695, respectively. In the external test cohort, the accuracy of the SE-ResNet50 model demonstrated a 10% improvement over the ResNet50 model (82.2% vs 72.2%). CONCLUSION The SE-ResNet50 model based on computed tomography super-resolution has great potential for predicting STAS status in patients with solid or partially solid LUAD, with superior predictive performance compared to traditional deep learning models.
Collapse
Affiliation(s)
- Shuxing Wang
- The Second Clinical Medical College, Jinan University, Shenzhen, Guangdong, China (S.W., X.L., Y.P., J.G.)
| | - Xiaowen Liu
- The Second Clinical Medical College, Jinan University, Shenzhen, Guangdong, China (S.W., X.L., Y.P., J.G.)
| | - Changsi Jiang
- Department of Radiology, Shenzhen People's Hospital (The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology), Floor 1 Bldg 4, Dongbeilu 1017, Shenzhen 518020, Guangdong, China (C.J., X.T., Y.L., J.G.)
| | - Wenyan Kang
- Department of Radiology, Cancer Hospital Chinese Academy of Medical Sciences, Shenzhen Center, Shenzhen, China (W.K.)
| | - Yudie Pan
- The Second Clinical Medical College, Jinan University, Shenzhen, Guangdong, China (S.W., X.L., Y.P., J.G.)
| | - Xue Tang
- Department of Radiology, Shenzhen People's Hospital (The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology), Floor 1 Bldg 4, Dongbeilu 1017, Shenzhen 518020, Guangdong, China (C.J., X.T., Y.L., J.G.)
| | - Yan Luo
- Department of Radiology, Shenzhen People's Hospital (The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology), Floor 1 Bldg 4, Dongbeilu 1017, Shenzhen 518020, Guangdong, China (C.J., X.T., Y.L., J.G.)
| | - Jingshan Gong
- The Second Clinical Medical College, Jinan University, Shenzhen, Guangdong, China (S.W., X.L., Y.P., J.G.); Department of Radiology, Shenzhen People's Hospital (The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology), Floor 1 Bldg 4, Dongbeilu 1017, Shenzhen 518020, Guangdong, China (C.J., X.T., Y.L., J.G.).
| |
Collapse
|
48
|
Kersch CN, Kim M, Stoller J, Barajas RF, Park JE. Imaging Genomics of Glioma Revisited: Analytic Methods to Understand Spatial and Temporal Heterogeneity. AJNR Am J Neuroradiol 2024; 45:537-548. [PMID: 38548303 PMCID: PMC11288537 DOI: 10.3174/ajnr.a8148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 11/09/2023] [Indexed: 04/12/2024]
Abstract
An improved understanding of the cellular and molecular biologic processes responsible for brain tumor development, growth, and resistance to therapy is fundamental to improving clinical outcomes. Imaging genomics is the study of the relationships between microscopic, genetic, and molecular biologic features and macroscopic imaging features. Imaging genomics is beginning to shift clinical paradigms for diagnosing and treating brain tumors. This article provides an overview of imaging genomics in gliomas, in which imaging data including hallmarks such as IDH-mutation, MGMT methylation, and EGFR-mutation status can provide critical insights into the pretreatment and posttreatment stages. This article will accomplish the following: 1) review the methods used in imaging genomics, including visual analysis, quantitative analysis, and radiomics analysis; 2) recommend suitable analytic methods for imaging genomics according to biologic characteristics; 3) discuss the clinical applicability of imaging genomics; and 4) introduce subregional tumor habitat analysis with the goal of guiding future radiogenetics research endeavors toward translation into critically needed clinical applications.
Collapse
Affiliation(s)
- Cymon N Kersch
- From the Department of Radiation Medicine (C.N.K.), Oregon Health and Science University, Portland, Oregon
| | - Minjae Kim
- Department of Radiology and Research Institute of Radiology (M.K., J.E.P.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jared Stoller
- Department of Diagnostic Radiology (J.S., R.F.B.), Oregon Health and Science University, Portland, Oregon
| | - Ramon F Barajas
- Department of Diagnostic Radiology (J.S., R.F.B.), Oregon Health and Science University, Portland, Oregon
- Knight Cancer Institute (R.F.B.), Oregon Health and Science University, Portland, Oregon
- Advanced Imaging Research Center (R.F.B.), Oregon Health and Science University, Portland, Oregon
| | - Ji Eun Park
- Department of Radiology and Research Institute of Radiology (M.K., J.E.P.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
49
|
Yuan J, Siakallis L, Li HB, Brandner S, Zhang J, Li C, Mancini L, Bisdas S. Structural- and DTI- MRI enable automated prediction of IDH Mutation Status in CNS WHO Grade 2-4 glioma patients: a deep Radiomics Approach. BMC Med Imaging 2024; 24:104. [PMID: 38702613 PMCID: PMC11067215 DOI: 10.1186/s12880-024-01274-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 04/15/2024] [Indexed: 05/06/2024] Open
Abstract
BACKGROUND The role of isocitrate dehydrogenase (IDH) mutation status for glioma stratification and prognosis is established. While structural magnetic resonance image (MRI) is a promising biomarker, it may not be sufficient for non-invasive characterisation of IDH mutation status. We investigated the diagnostic value of combined diffusion tensor imaging (DTI) and structural MRI enhanced by a deep radiomics approach based on convolutional neural networks (CNNs) and support vector machine (SVM), to determine the IDH mutation status in Central Nervous System World Health Organization (CNS WHO) grade 2-4 gliomas. METHODS This retrospective study analyzed the DTI-derived fractional anisotropy (FA) and mean diffusivity (MD) images and structural images including fluid attenuated inversion recovery (FLAIR), non-enhanced T1-, and T2-weighted images of 206 treatment-naïve gliomas, including 146 IDH mutant and 60 IDH-wildtype ones. The lesions were manually segmented by experienced neuroradiologists and the masks were applied to the FA and MD maps. Deep radiomics features were extracted from each subject by applying a pre-trained CNN and statistical description. An SVM classifier was applied to predict IDH status using imaging features in combination with demographic data. RESULTS We comparatively assessed the CNN-SVM classifier performance in predicting IDH mutation status using standalone and combined structural and DTI-based imaging features. Combined imaging features surpassed stand-alone modalities for the prediction of IDH mutation status [area under the curve (AUC) = 0.846; sensitivity = 0.925; and specificity = 0.567]. Importantly, optimal model performance was noted following the addition of demographic data (patients' age) to structural and DTI imaging features [area under the curve (AUC) = 0.847; sensitivity = 0.911; and specificity = 0.617]. CONCLUSIONS Imaging features derived from DTI-based FA and MD maps combined with structural MRI, have superior diagnostic value to that provided by standalone structural or DTI sequences. In combination with demographic information, this CNN-SVM model offers a further enhanced non-invasive prediction of IDH mutation status in gliomas.
Collapse
Affiliation(s)
- Jialin Yuan
- Department of Radiology, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
- Queen Square Institute of Neurology, University College London, London, UK
| | - Loizos Siakallis
- Queen Square Institute of Neurology, University College London, London, UK
| | - Hongwei Bran Li
- Department of Informatics, Technical University of Munich, Munich, Germany
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, USA
| | - Sebastian Brandner
- Division of Neuropathology, Queen Square Institute of Neurology, University College London, London, UK
| | - Jianguo Zhang
- Department of Computer Science and Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Chenming Li
- Department of Computer Science and Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Laura Mancini
- Queen Square Institute of Neurology, University College London, London, UK
- Lysholm Department of Neuroradiology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Sotirios Bisdas
- Queen Square Institute of Neurology, University College London, London, UK.
- Lysholm Department of Neuroradiology, University College London Hospitals NHS Foundation Trust, London, UK.
| |
Collapse
|
50
|
Gao J, Liu Z, Pan H, Cao X, Kan Y, Wen Z, Chen S, Wen M, Zhang L. Preoperative Discrimination of CDKN2A/B Homozygous Deletion Status in Isocitrate Dehydrogenase-Mutant Astrocytoma: A Deep Learning-Based Radiomics Model Using MRI. J Magn Reson Imaging 2024; 59:1655-1664. [PMID: 37555723 DOI: 10.1002/jmri.28945] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 07/26/2023] [Accepted: 07/26/2023] [Indexed: 08/10/2023] Open
Abstract
BACKGROUND Cyclin-dependent kinase inhibitor 2A/B (CDKN2A/B) homozygous deletion has been verified as an independent and critical biomarker of negative prognosis and short survival in isocitrate dehydrogenase (IDH)-mutant astrocytoma. Therefore, noninvasive and accurate discrimination of CDKN2A/B homozygous deletion status is essential for the clinical management of IDH-mutant astrocytoma patients. PURPOSE To develop a noninvasive, robust preoperative model based on MR image features for discriminating CDKN2A/B homozygous deletion status of IDH-mutant astrocytoma. STUDY TYPE Retrospective. POPULATION Two hundred fifty-one patients: 107 patients with CDKN2A/B homozygous deletion and 144 patients without CDKN2A/B homozygous deletion. FIELD STRENGTH/SEQUENCE 3.0 T/1.5 T: Contrast-enhanced T1-weighted spin-echo inversion recovery sequence (CE-T1WI) and T2-weighted fluid-attenuation spin-echo inversion recovery sequence (T2FLAIR). ASSESSMENT A total of 1106 radiomics and 1000 deep learning features extracted from CE-T1WI and T2FLAIR were used to develop models to discriminate the CDKN2A/B homozygous deletion status. Radiomics models, deep learning-based radiomics (DLR) models and the final integrated model combining radiomics features with deep learning features were developed and compared their preoperative discrimination performance. STATISTICAL TESTING Pearson chi-square test and Mann Whitney U test were used for assessing the statistical differences in patients' clinical characteristics. The Delong test compared the statistical differences of receiver operating characteristic (ROC) curves and area under the curve (AUC) of different models. The significance threshold is P < 0.05. RESULTS The final combined model (training AUC = 0.966; validation AUC = 0.935; test group: AUC = 0.943) outperformed the optimal models based on only radiomics or DLR features (training: AUC = 0.916 and 0.952; validation: AUC = 0.886 and 0.912; test group: AUC = 0.862 and 0.902). DATA CONCLUSION Whether based on a single sequence or a combination of two sequences, radiomics and DLR models have achieved promising performance in assessing CDKN2A/B homozygous deletion status. However, the final model combining both deep learning and radiomics features from CE-T1WI and T2FLAIR outperformed the optimal radiomics or DLR model. EVIDENCE LEVEL 4 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Jueni Gao
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhi Liu
- Department of Nuclear Medicine, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Hongyu Pan
- College of Computer and Information Science, Southwest University, Chongqing, China
| | - Xu Cao
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yubo Kan
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhipeng Wen
- Department of Radiology, Sichuan Cancer Hospital, Chengdu, China
| | - Shanxiong Chen
- College of Computer and Information Science, Southwest University, Chongqing, China
| | - Ming Wen
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Liqiang Zhang
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|