1
|
Mohammadpour S, Emami H, Rabiei R, Hosseini A, Moghaddasi H, Faeghi F, Bagherzadeh R. Image Analysis as tool for Predicting Colorectal Cancer Molecular Alterations: A Scoping Review. Mol Imaging Radionucl Ther 2025; 34:10-25. [PMID: 39917985 PMCID: PMC11827529 DOI: 10.4274/mirt.galenos.2024.86402] [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: 05/06/2024] [Accepted: 08/25/2024] [Indexed: 02/16/2025] Open
Abstract
Objectives Among the most important diagnostic indicators of colorectal cancer; however, measuring molecular alterations are invasive and expensive. This study aimed to investigate the application of image processing to predict molecular alterations in colorectal cancer. Methods In this scoping review, we searched for relevant literature by searching the Web of Science, Scopus, and PubMed databases. The method of selecting the articles and reporting the findings was according to the guidelines of the Preferred Reporting Items for Systematic Reviews and Meta-Analyses; moreover, the Strengthening the Reporting of Observational Studies in Epidemiology checklist was used to assess the quality of the studies. Results Sixty seven out of 2,223 articles, 67 were relevant to the aim of the study, and finally 41 studies with sufficient quality were reviewed. The prediction of Kirsten Rat Sarcoma Viral Oncogene Homolog (KRAS), Neuroblastoma RAS Viral (NRAS), B-Raf proto-oncogene, serine/threonine kinase (BRAF), Tumor Protein 53 (TP53), Adenomatous Polyposis Coli, and microsatellite instability (MSI) with the help of image analysis has received more attention than other molecular characteristics. The studies used computed tomography (CT), magnetic resonance imaging (MRI), and 18F-FDG positron emission tomography (PET)/CT with radionics and quantitative analysis to predict molecular alterations in colorectal cancer, analyzing features like texture, maximum standard uptake value, and MTV using various statistical methods. In 39 studies, there was a significant relationship between the features extracted from these images and molecular alterations. Different modalities were used to measure the area under the receiver operating characteristic curve for predicting the alterations in KRAS, MSI, BRAF, and TP53, with an average of 78, 81, 80 and 71%, respectively. Conclusion This scoping review underscores the potential of radiogenomics in predicting molecular alterations in colorectal cancer through non-invasive imaging modalities, like CT, MRI, and 18F-FDG PET/CT. The analysis of 41 studies showed the appropriate prediction of key alterations, such as KRAS, NRAS, BRAF, TP53, and MSI, highlighting the promise of radionics and texture features in enhancing predictive accuracy.
Collapse
Affiliation(s)
- Saman Mohammadpour
- Shahid Beheshti University Faculty of Medicine, Department of Health Information Technology and Management, Tehran, Iran
| | - Hassan Emami
- Shahid Beheshti University Faculty of Medicine, Department of Health Information Technology and Management, Tehran, Iran
| | - Reza Rabiei
- Shahid Beheshti University Faculty of Medicine, Department of Health Information Technology and Management, Tehran, Iran
| | - Azamossadat Hosseini
- Shahid Beheshti University Faculty of Medicine, Department of Health Information Technology and Management, Tehran, Iran
| | - Hamid Moghaddasi
- Shahid Beheshti University Faculty of Medicine, Department of Health Information Technology and Management, Tehran, Iran
| | - Fariborz Faeghi
- Shahid Beheshti University Faculty of Medicine, Department of Radiology Technology, Tehran, Iran
| | - Rafat Bagherzadeh
- Iran University of Medical Sciences Faculty of Medicine, Department of English Language, Tehran, Iran
| |
Collapse
|
2
|
Guo Y, Li T, Gong B, Hu Y, Wang S, Yang L, Zheng C. From Images to Genes: Radiogenomics Based on Artificial Intelligence to Achieve Non-Invasive Precision Medicine in Cancer Patients. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2408069. [PMID: 39535476 PMCID: PMC11727298 DOI: 10.1002/advs.202408069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 10/19/2024] [Indexed: 11/16/2024]
Abstract
With the increasing demand for precision medicine in cancer patients, radiogenomics emerges as a promising frontier. Radiogenomics is originally defined as a methodology for associating gene expression information from high-throughput technologies with imaging phenotypes. However, with advancements in medical imaging, high-throughput omics technologies, and artificial intelligence, both the concept and application of radiogenomics have significantly broadened. In this review, the history of radiogenomics is enumerated, related omics technologies, the five basic workflows and their applications across tumors, the role of AI in radiogenomics, the opportunities and challenges from tumor heterogeneity, and the applications of radiogenomics in tumor immune microenvironment. The application of radiogenomics in positron emission tomography and the role of radiogenomics in multi-omics studies is also discussed. Finally, the challenges faced by clinical transformation, along with future trends in this field is discussed.
Collapse
Affiliation(s)
- Yusheng Guo
- Department of RadiologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Hubei Key Laboratory of Molecular ImagingWuhan430022China
| | - Tianxiang Li
- Department of UltrasoundState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical. SciencesPeking Union Medical CollegeBeijing100730China
| | - Bingxin Gong
- Department of RadiologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Hubei Key Laboratory of Molecular ImagingWuhan430022China
| | - Yan Hu
- Research Institute of Trustworthy Autonomous Systems and Department of Computer Science and EngineeringSouthern University of Science and TechnologyShenzhen518055China
| | - Sichen Wang
- School of Life Science and TechnologyComputational Biology Research CenterHarbin Institute of TechnologyHarbin150001China
| | - Lian Yang
- Department of RadiologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Hubei Key Laboratory of Molecular ImagingWuhan430022China
| | - Chuansheng Zheng
- Department of RadiologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Hubei Key Laboratory of Molecular ImagingWuhan430022China
| |
Collapse
|
3
|
Li Z, Zhang J, Zhong Q, Feng Z, Shi Y, Xu L, Zhang R, Yu F, Lv B, Yang T, Huang C, Cui F, Chen F. Development and external validation of a multiparametric MRI-based radiomics model for preoperative prediction of microsatellite instability status in rectal cancer: a retrospective multicenter study. Eur Radiol 2023; 33:1835-1843. [PMID: 36282309 DOI: 10.1007/s00330-022-09160-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 07/27/2022] [Accepted: 08/29/2022] [Indexed: 11/30/2022]
Abstract
OBJECTIVES To establish and validate a radiomics model based on multiparametric magnetic resonance imaging (MRI), and to predict microsatellite instability (MSI) status in rectal cancer patients. METHODS A total of 199 patients with pathologically confirmed rectal cancer were included. The MSI status was confirmed by immunohistochemistry (IHC) staining. Clinical factors and laboratory data associated with MSI status were analyzed. The imaging data of 100 patients from one of the hospitals were used as the training set. The remaining 99 patients from the other two hospitals were used as the external validation set. The regions of interest (ROIs) were delineated from T1-weighted imaging (T1WI), T2-weighted imaging (T2WI), diffusion-weighted imaging (DWI), and contrast-enhanced T1WI (CE-T1WI) sequence to extract the radiomics features. The Tree-based approach was used for feature selection. The models were constructed based on the four single sequences and a combination of the four sequences using the random forest (RF) algorithm. The external validation set was used to verify the generalization ability of each model. The receiver operating characteristic (ROC) curves and the area under the curve (AUC) were plotted to evaluate and compare the predictive performance of each model. RESULTS In the four single-series models, the CE-T1WI model performed the best. The AUCs of the T1WI, T2WI, DWI, and CE-T1WI prediction models in the training set were 0.74, 0.71, 0.71, and 0.78, respectively, while in the external validation set, the corresponding AUCs were 0.67, 0.66, 0.70, and 0.77. The prediction and generalization performance of the combined model of multi-sequences was comparable to that of the CE-T1WI model and it was better than that of the remaining three single-series models, with AUC values of 0.78 and 0.78 in the training and validation sets, respectively. CONCLUSION The established radiomics models based on CE-T1WI or multiparametric MRI have similar predictive performance. They have the potential to predict MSI status in rectal cancer patients. KEY POINTS • A radiomics model for the prediction of MSI status in patients with rectal cancer was established and validated using external validation. • The models based on CE-T1WI or multiparametric MRI have better predictive performance than those based on single unenhanced sequence images. • The radiomics model has the potential to suggest MSI status in rectal cancer patients; however, it is not yet a substitute for histological confirmation.
Collapse
Affiliation(s)
- Zhi Li
- Department of Radiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jing Zhang
- Department of Radiology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qi Zhong
- Department of Radiology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, China
| | - Zhan Feng
- Department of Radiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yushu Shi
- Department of Radiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ligong Xu
- Department of Radiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Rui Zhang
- Department of Radiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Fang Yu
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Biao Lv
- Department of Radiology, The 903 Hospital of Joint Logistics Support Force of PLA, Hangzhou, Zhejiang, China
| | - Tian Yang
- Department of Radiology, Shulan (Hangzhou) Hospital, Hangzhou, Zhejiang, China
| | - Chencui Huang
- Department of Research Collaboration, R&D Center, Beijing Deepwise & League of PHD Technology Co., Ltd, Beijing, China
| | - Feng Cui
- Department of Radiology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, China
| | - Feng Chen
- Department of Radiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
4
|
Zhang S, Mu W, Dong D, Wei J, Fang M, Shao L, Zhou Y, He B, Zhang S, Liu Z, Liu J, Tian J. The Applications of Artificial Intelligence in Digestive System Neoplasms: A Review. HEALTH DATA SCIENCE 2023; 3:0005. [PMID: 38487199 PMCID: PMC10877701 DOI: 10.34133/hds.0005] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 12/05/2022] [Indexed: 03/17/2024]
Abstract
Importance Digestive system neoplasms (DSNs) are the leading cause of cancer-related mortality with a 5-year survival rate of less than 20%. Subjective evaluation of medical images including endoscopic images, whole slide images, computed tomography images, and magnetic resonance images plays a vital role in the clinical practice of DSNs, but with limited performance and increased workload of radiologists or pathologists. The application of artificial intelligence (AI) in medical image analysis holds promise to augment the visual interpretation of medical images, which could not only automate the complicated evaluation process but also convert medical images into quantitative imaging features that associated with tumor heterogeneity. Highlights We briefly introduce the methodology of AI for medical image analysis and then review its clinical applications including clinical auxiliary diagnosis, assessment of treatment response, and prognosis prediction on 4 typical DSNs including esophageal cancer, gastric cancer, colorectal cancer, and hepatocellular carcinoma. Conclusion AI technology has great potential in supporting the clinical diagnosis and treatment decision-making of DSNs. Several technical issues should be overcome before its application into clinical practice of DSNs.
Collapse
Affiliation(s)
- Shuaitong Zhang
- School of Engineering Medicine, Beihang University, Beijing, China
- Key Laboratory of Big Data-Based Precision Medicine, Beihang University, Ministry of Industry and Information Technology, Beijing, China
| | - Wei Mu
- School of Engineering Medicine, Beihang University, Beijing, China
- Key Laboratory of Big Data-Based Precision Medicine, Beihang University, Ministry of Industry and Information Technology, Beijing, China
| | - Di Dong
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Jingwei Wei
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Mengjie Fang
- School of Engineering Medicine, Beihang University, Beijing, China
- Key Laboratory of Big Data-Based Precision Medicine, Beihang University, Ministry of Industry and Information Technology, Beijing, China
| | - Lizhi Shao
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Yu Zhou
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Bingxi He
- School of Engineering Medicine, Beihang University, Beijing, China
- Key Laboratory of Big Data-Based Precision Medicine, Beihang University, Ministry of Industry and Information Technology, Beijing, China
| | - Song Zhang
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Zhenyu Liu
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Jianhua Liu
- Department of Oncology, Guangdong Provincial People's Hospital/Second Clinical Medical College of Southern Medical University/Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Jie Tian
- School of Engineering Medicine, Beihang University, Beijing, China
- Key Laboratory of Big Data-Based Precision Medicine, Beihang University, Ministry of Industry and Information Technology, Beijing, China
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
5
|
Ghaffari Laleh N, Ligero M, Perez-Lopez R, Kather JN. Facts and Hopes on the Use of Artificial Intelligence for Predictive Immunotherapy Biomarkers in Cancer. Clin Cancer Res 2023; 29:316-323. [PMID: 36083132 DOI: 10.1158/1078-0432.ccr-22-0390] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/26/2022] [Accepted: 08/29/2022] [Indexed: 01/19/2023]
Abstract
Immunotherapy by immune checkpoint inhibitors has become a standard treatment strategy for many types of solid tumors. However, the majority of patients with cancer will not respond, and predicting response to this therapy is still a challenge. Artificial intelligence (AI) methods can extract meaningful information from complex data, such as image data. In clinical routine, radiology or histopathology images are ubiquitously available. AI has been used to predict the response to immunotherapy from radiology or histopathology images, either directly or indirectly via surrogate markers. While none of these methods are currently used in clinical routine, academic and commercial developments are pointing toward potential clinical adoption in the near future. Here, we summarize the state of the art in AI-based image biomarkers for immunotherapy response based on radiology and histopathology images. We point out limitations, caveats, and pitfalls, including biases, generalizability, and explainability, which are relevant for researchers and health care providers alike, and outline key clinical use cases of this new class of predictive biomarkers.
Collapse
Affiliation(s)
| | - Marta Ligero
- Radiomics Group, Vall d'Hebron Institute of Oncology, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Raquel Perez-Lopez
- Radiomics Group, Vall d'Hebron Institute of Oncology, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain.,Department of Radiology, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Jakob Nikolas Kather
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany.,Division of Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, United Kingdom.,Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany.,Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| |
Collapse
|
6
|
Zhang Y, Liu J, Wu C, Peng J, Wei Y, Cui S. Preoperative Prediction of Microsatellite Instability in Rectal Cancer Using Five Machine Learning Algorithms Based on Multiparametric MRI Radiomics. Diagnostics (Basel) 2023; 13:diagnostics13020269. [PMID: 36673079 PMCID: PMC9858257 DOI: 10.3390/diagnostics13020269] [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: 10/11/2022] [Revised: 12/29/2022] [Accepted: 01/05/2023] [Indexed: 01/13/2023] Open
Abstract
Objectives: To establish and verify radiomics models based on multiparametric MRI for preoperatively identifying the microsatellite instability (MSI) status of rectal cancer (RC) by comparing different machine learning algorithms. Methods: This retrospective study enrolled 383 (training set, 268; test set, 115) RC patients between January 2017 and June 2022. A total of 4148 radiomics features were extracted from multiparametric MRI, including T2-weighted imaging, T1-weighted imaging, apparent diffusion coefficient, and contrast-enhanced T1-weighted imaging. The analysis of variance, correlation test, univariate logistic analysis, and a gradient-boosting decision tree were used for the dimension reduction. Logistic regression, Bayes, support vector machine (SVM), K-nearest neighbor (KNN), and tree machine learning algorithms were used to build different radiomics models. The relative standard deviation (RSD) and bootstrap method were used to quantify the stability of these five algorithms. Then, predictive performances of different models were assessed using area under curves (AUCs). The performance of the best radiomics model was evaluated using calibration and discrimination. Results: Among these 383 patients, the prevalence of MSI was 14.62% (56/383). The RSD value of logistic regression algorithm was the lowest (4.64%), followed by Bayes (5.44%) and KNN (5.45%), which was significantly better than that of SVM (19.11%) and tree (11.94%) algorithms. The radiomics model based on logistic regression algorithm performed best, with AUCs of 0.827 and 0.739 in the training and test sets, respectively. Conclusions: We developed a radiomics model based on the logistic regression algorithm, which could potentially be used to facilitate the individualized prediction of MSI status in RC patients.
Collapse
Affiliation(s)
- Yang Zhang
- Cancer Center, Department of Radiology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou 310014, China
| | - Jing Liu
- Cancer Center, Department of Radiology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou 310014, China
| | - Cuiyun Wu
- Cancer Center, Department of Radiology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou 310014, China
| | - Jiaxuan Peng
- Medical College, Jinzhou Medical University, Jinzhou 121001, China
| | - Yuguo Wei
- Precision Health Institution, General Electric Healthcare, Hangzhou 310004, China
| | - Sijia Cui
- Cancer Center, Department of Radiology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou 310014, China
- Correspondence:
| |
Collapse
|
7
|
Zhou H, Luo Q, Wu W, Li N, Yang C, Zou L. Radiomics-guided checkpoint inhibitor immunotherapy for precision medicine in cancer: A review for clinicians. Front Immunol 2023; 14:1088874. [PMID: 36936913 PMCID: PMC10014595 DOI: 10.3389/fimmu.2023.1088874] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 02/16/2023] [Indexed: 03/05/2023] Open
Abstract
Immunotherapy using immune checkpoint inhibitors (ICIs) is a breakthrough in oncology development and has been applied to multiple solid tumors. However, unlike traditional cancer treatment approaches, immune checkpoint inhibitors (ICIs) initiate indirect cytotoxicity by generating inflammation, which causes enlargement of the lesion in some cases. Therefore, rather than declaring progressive disease (PD) immediately, confirmation upon follow-up radiological evaluation after four-eight weeks is suggested according to immune-related Response Evaluation Criteria in Solid Tumors (ir-RECIST). Given the difficulty for clinicians to immediately distinguish pseudoprogression from true disease progression, we need novel tools to assist in this field. Radiomics, an innovative data analysis technique that quantifies tumor characteristics through high-throughput extraction of quantitative features from images, can enable the detection of additional information from early imaging. This review will summarize the recent advances in radiomics concerning immunotherapy. Notably, we will discuss the potential of applying radiomics to differentiate pseudoprogression from PD to avoid condition exacerbation during confirmatory periods. We also review the applications of radiomics in hyperprogression, immune-related biomarkers, efficacy, and immune-related adverse events (irAEs). We found that radiomics has shown promising results in precision cancer immunotherapy with early detection in noninvasive ways.
Collapse
Affiliation(s)
- Huijie Zhou
- Division of Medical Oncology, Cancer Center and State Key Laboratory of Biotherapy, Sichuan University West China Hospital, Chengdu, China
| | - Qian Luo
- Department of Hematology, the Second Affiliated Hospital Zhejiang University School of Medicine, Zhejiang, China
| | - Wanchun Wu
- Division of Medical Oncology, Cancer Center and State Key Laboratory of Biotherapy, Sichuan University West China Hospital, Chengdu, China
| | - Na Li
- Division of Medical Oncology, Cancer Center and State Key Laboratory of Biotherapy, Sichuan University West China Hospital, Chengdu, China
| | - Chunli Yang
- Division of Medical Oncology, Cancer Center and State Key Laboratory of Biotherapy, Sichuan University West China Hospital, Chengdu, China
| | - Liqun Zou
- Division of Medical Oncology, Cancer Center and State Key Laboratory of Biotherapy, Sichuan University West China Hospital, Chengdu, China
- *Correspondence: Liqun Zou,
| |
Collapse
|
8
|
Chen X, He L, Li Q, Liu L, Li S, Zhang Y, Liu Z, Huang Y, Mao Y, Chen X. Non-invasive prediction of microsatellite instability in colorectal cancer by a genetic algorithm-enhanced artificial neural network-based CT radiomics signature. Eur Radiol 2023; 33:11-22. [PMID: 35771245 DOI: 10.1007/s00330-022-08954-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 05/08/2022] [Accepted: 06/08/2022] [Indexed: 11/25/2022]
Abstract
OBJECTIVE The stratification of microsatellite instability (MSI) status assists clinicians in making treatment decisions for colorectal cancer (CRC) patients. This study aimed to establish a CT-based radiomics signature to predict MSI status in patients with CRC. METHODS A total of 837 CRC patients who underwent preoperative enhanced CT and had available MSI status data were recruited from two hospitals. Radiomics features were extracted from segmented tumours, and a series of data balancing and feature selection strategies were used to select MSI-related features. Finally, an MSI-related radiomics signature was constructed using a genetic algorithm-enhanced artificial neural network model. Combined and clinical models were constructed using multivariate logistic regression analyses by integrating the clinical factors with or without the signature. A Kaplan-Meier survival analysis was conducted to explore the prognostic information of the signature in patients with CRC. RESULTS Ten features were selected to construct a signature which showed robust performance in both the internal and external validation cohorts, with areas under the curves (AUC) of 0.788 and 0.775, respectively. The performance of the signature was comparable to that of the combined model (AUCs of 0.777 and 0.767, respectively) and it outperformed the clinical model constituting age and tumour location (AUCs of 0.768 and 0.623, respectively). Survival analysis demonstrated that the signature could stratify patients with stage II CRC according to prognosis (HR: 0.402, p = 0.029). CONCLUSIONS This study built a robust radiomics signature for identifying the MSI status of CRC patients, which may assist individualised treatment decisions. KEY POINTS • Our well-designed modelling strategies helped overcome the problem of data imbalance caused by the low incidence of MSI. • Genetic algorithm-enhanced artificial neural network-based CT radiomics signature can effectively distinguish the MSI status of CRC patients. • Kaplan-Meier survival analysis demonstrated that our signature could significantly stratify stage II CRC patients into high- and low-risk groups.
Collapse
Affiliation(s)
- Xiaobo Chen
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Lan He
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Qingshu Li
- Department of Pathology, College of Basic Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Liu Liu
- Department of Radiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Suyun Li
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
- School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Yuan Zhang
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Zaiyi Liu
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Yanqi Huang
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, China.
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China.
| | - Yun Mao
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, China.
- Department of Radiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Xin Chen
- Department of Radiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, 1 Panfu Road, Guangzhou, 510180, China.
| |
Collapse
|
9
|
Ma Y, Lin C, Liu S, Wei Y, Ji C, Shi F, Lin F, Zhou Z. Radiomics features based on internal and marginal areas of the tumor for the preoperative prediction of microsatellite instability status in colorectal cancer. Front Oncol 2022; 12:1020349. [PMID: 36276101 PMCID: PMC9583004 DOI: 10.3389/fonc.2022.1020349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 09/20/2022] [Indexed: 11/21/2022] Open
Abstract
Objectives To explore whether the preoperative CT radiomics can predict the status of microsatellite instability (MSI) in colorectal cancer (CRC) patients and identify the region with the most stable and high-efficiency radiomics features. Methods This retrospective study involved 230 CRC patients with preoperative computed tomography scans and available MSI status between December 2019 and October 2021. Image segmentation and radiomic feature extraction were performed as follows. First, slices with the maximum tumor area (region of interest, ROI) were manually contoured. Subsequently, each ROI was shrunk inward by 1, 2, and 3 mm, respectively, where the remaining ROIs were considered as the internal region of the tumor (named as IROI1, IROI2, and IROI3), and the shrunk regions were considered as marginal regions of the tumor (named as MROI1, MROI2, and MROI3). Finally, radiomics features were extracted from each of the ROI. The intraclass correlation coefficient and least absolute shrinkage and selection operator method were used to choose the most reliable and relevant features of MSI status. Clinical, radiomics, and combined clinical radiomics models have been established. Calibration curve and decision curve analyses (DCA) were generated to explore the correction effect and assess the clinical applicability of the above models, respectively. Results In the testing cohort, the radiomics model based on IROI3 yielded the highest average area under the curve (AUC) value of 0.908, compared with the remaining radiomics models. Additionally, hypertension and N stage were considered as clinically independent factors of MSI status. The combined clinical radiomics model achieved excellent diagnostic efficacy (AUC: 0.928; sensitivity: 0.840; specificity: 0.867) in the testing cohort, as well as favorable calibration and clinical utility by calibration curve and DCA analyses. Conclusions The IROI3 model, which is based on a 3-mm shrink in the largest areas of the tumor, could noninvasively reflect the heterogeneity and genetic instability within the tumor. This suggests that it is an important biomarker for the preoperative prediction of MSI status. The model can extract more robust and effective radiomics features, which lays a foundation for the radiomics study of hollow organs, such as in CRC.
Collapse
Affiliation(s)
- Yi Ma
- Department of Radiology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Changsong Lin
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
| | - Song Liu
- Department of Radiology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Ying Wei
- Department of Research and Development, Shanghai United Imaging Intelligence Co., Ltd., Shanghai, China
| | - Changfeng Ji
- Department of Radiology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Feng Shi
- Department of Research and Development, Shanghai United Imaging Intelligence Co., Ltd., Shanghai, China
| | - Fan Lin
- Department of Cell Biology, Nanjing Medical University, Nanjing, China
- *Correspondence: Fan Lin, ; Zhengyang Zhou,
| | - Zhengyang Zhou
- Department of Radiology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
- *Correspondence: Fan Lin, ; Zhengyang Zhou,
| |
Collapse
|
10
|
Zeng Q, Zhu Y, Li L, Feng Z, Shu X, Wu A, Luo L, Cao Y, Tu Y, Xiong J, Zhou F, Li Z. CT-based radiomic nomogram for preoperative prediction of DNA mismatch repair deficiency in gastric cancer. Front Oncol 2022; 12:883109. [PMID: 36185292 PMCID: PMC9523515 DOI: 10.3389/fonc.2022.883109] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundDNA mismatch repair (MMR) deficiency has attracted considerable attention as a predictor of the immunotherapy efficacy of solid tumors, including gastric cancer. We aimed to develop and validate a computed tomography (CT)-based radiomic nomogram for the preoperative prediction of MMR deficiency in gastric cancer (GC).MethodsIn this retrospective analysis, 225 and 91 GC patients from two distinct hospital cohorts were included. Cohort 1 was randomly divided into a training cohort (n = 176) and an internal validation cohort (n = 76), whereas cohort 2 was considered an external validation cohort. Based on repeatable radiomic features, a radiomic signature was constructed using the least absolute shrinkage and selection operator (LASSO) regression analysis. We employed multivariable logistic regression analysis to build a radiomics-based model based on radiomic features and preoperative clinical characteristics. Furthermore, this prediction model was presented as a radiomic nomogram, which was evaluated in the training, internal validation, and external validation cohorts.ResultsThe radiomic signature composed of 15 robust features showed a significant association with MMR protein status in the training, internal validation, and external validation cohorts (both P-values <0.001). A radiomic nomogram incorporating a radiomic signature and two clinical characteristics (age and CT-reported N stage) represented good discrimination in the training cohort with an AUC of 0.902 (95% CI: 0.853–0.951), in the internal validation cohort with an AUC of 0.972 (95% CI: 0.945–1.000) and in the external validation cohort with an AUC of 0.891 (95% CI: 0.825–0.958).ConclusionThe CT-based radiomic nomogram showed good performance for preoperative prediction of MMR protein status in GC. Furthermore, this model was a noninvasive tool to predict MMR protein status and guide neoadjuvant therapy.
Collapse
Affiliation(s)
- Qingwen Zeng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, China
- Institute of Digestive Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yanyan Zhu
- Department of Radiology, The First Affiliated Hospital, Nanchang University, Nanchang, China
| | - Leyan Li
- Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Zongfeng Feng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, China
- Institute of Digestive Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xufeng Shu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, China
| | - Ahao Wu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, China
| | - Lianghua Luo
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, China
| | - Yi Cao
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, China
- *Correspondence: Zhengrong Li, ; Yi Cao,
| | - Yi Tu
- Department of Pathology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jianbo Xiong
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, China
| | - Fuqing Zhou
- Department of Radiology, The First Affiliated Hospital, Nanchang University, Nanchang, China
| | - Zhengrong Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, China
- Institute of Digestive Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
- *Correspondence: Zhengrong Li, ; Yi Cao,
| |
Collapse
|
11
|
Detection of Microsatellite Instability in Colonoscopic Biopsies and Postal Urine Samples from Lynch Syndrome Cancer Patients Using a Multiplex PCR Assay. Cancers (Basel) 2022; 14:cancers14153838. [PMID: 35954501 PMCID: PMC9367254 DOI: 10.3390/cancers14153838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/15/2022] [Accepted: 07/21/2022] [Indexed: 11/16/2022] Open
Abstract
Identification of mismatch repair (MMR)-deficient colorectal cancers (CRCs) is recommended for Lynch syndrome (LS) screening, and supports targeting of immune checkpoint inhibitors. Microsatellite instability (MSI) analysis is commonly used to test for MMR deficiency. Testing biopsies prior to tumour resection can inform surgical and therapeutic decisions, but can be limited by DNA quantity. MSI analysis of voided urine could also provide much needed surveillance for genitourinary tract cancers in LS. Here, we reconfigure an existing molecular inversion probe-based MSI and BRAF c.1799T > A assay to a multiplex PCR (mPCR) format, and demonstrate that it can sample >140 unique molecules per marker from <1 ng of DNA and classify CRCs with 96−100% sensitivity and specificity. We also show that it can detect increased MSI within individual and composite CRC biopsies from LS patients, and within preoperative urine cell free DNA (cfDNA) from two LS patients, one with an upper tract urothelial cancer, the other an undiagnosed endometrial cancer. Approximately 60−70% of the urine cfDNAs were tumour-derived. Our results suggest that mPCR sequence-based analysis of MSI and mutation hotspots in CRC biopsies could facilitate presurgery decision making, and could enable postal-based screening for urinary tract and endometrial tumours in LS patients.
Collapse
|
12
|
A multicenter study on the preoperative prediction of gastric cancer microsatellite instability status based on computed tomography radiomics. Abdom Radiol (NY) 2022; 47:2036-2045. [PMID: 35391567 DOI: 10.1007/s00261-022-03507-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/21/2022] [Accepted: 03/22/2022] [Indexed: 12/16/2022]
Abstract
PURPOSE To construct and validate a radiomics feature model based on computed tomography (CT) images and clinical characteristics to predict the microsatellite instability (MSI) status of gastric cancer patients before surgery. METHODS We retrospectively collected the upper abdominal or the entire abdominal-enhanced CT scans of 189 gastric cancer patients before surgery. The patients underwent postoperative gastric cancer MSI status testing, and the dates of their radiologic images and clinicopathological data were from January 2015 to August 2021. These 189 patients were divided into a training set (n = 90) and an external validation set (n = 99). The patients were divided by MSI status into the MSI-high (H) arm (30 and 33 patients in the training set and external validation set, respectively) and MSI-low/stable (L/S) arm (60 and 66 patients in the training set and external validation set, respectively). In the training set, the clinical characteristics and tumor radiologic characteristics of the patients were extracted, and the tenfold cross-validation method was used for internal validation of the training set. The external validation set was used to assess its generalized performance. A receiver-operating characteristic (ROC) curve was plotted to assess the model performance, and the area under the curve (AUC) was calculated. RESULTS The AUC of the radiomics model in the training set and external validation set was 0.8228 [95% confidence interval (CI) 0.7355-0.9101] and 0.7603 [95% CI 0.6625-0.8581], respectively, showing that the constructed radiomics model exhibited satisfactory generalization capabilities. The accuracy, sensitivity, and specificity of the training dataset were 0.72, 0.63, and 0.77, respectively. The accuracy, sensitivity, and specificity of the external validation dataset were 0.67, 0.79, and 0.60, respectively. Statistical analysis was carried out on the clinical data, and there was statistical significance for the tumor site and age (p < 0.05). MSI-H gastric cancer was mostly seen in the gastric antrum and older patients. CONCLUSIONS Radiomics markers based on CT images and clinical characteristics have the potential to be a non-invasive auxiliary diagnostic tool for preoperative assessment of gastric cancer MSI status, and they can aid in clinical decision-making and improve patient outcomes.
Collapse
|
13
|
Multi-Omic Approaches in Colorectal Cancer beyond Genomic Data. J Pers Med 2022; 12:jpm12020128. [PMID: 35207616 PMCID: PMC8880341 DOI: 10.3390/jpm12020128] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/10/2022] [Accepted: 01/12/2022] [Indexed: 02/04/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most frequent tumours and one of the major causes of morbidity and mortality globally. Its incidence has increased in recent years and could be linked to unhealthy dietary habits combined with environmental and hereditary factors, which can lead to genetic and epigenetic changes and induce tumour development. The model of CRC progression has always been based on a genomic, parametric, static and complex approach involving oncogenes and tumour suppressor genes. Recent advances in omics sciences have sought a paradigm shift to a multiparametric, immunological-stromal, and dynamic approach for a better understanding of carcinogenesis and tumour heterogeneity. In the present paper, we review the most important preclinical and clinical data and present recent discoveries in the field of transcriptomics, proteomics, metagenomics and radiomics in CRC disease.
Collapse
|