151
|
Tolani MA, Zubairu IH, Balarabe K, Awaisu M, Abdullahi M, Adeniji AA, Umar SS, Bello A, Tagawa ST. Barriers and facilitators of the application of precision medicine to the genitourinary cancer care pathway: Perspective from a low- and middle- income country in sub-Saharan Africa. Urol Oncol 2024; 42:411-420. [PMID: 39183140 DOI: 10.1016/j.urolonc.2024.07.014] [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: 01/19/2024] [Revised: 07/13/2024] [Accepted: 07/19/2024] [Indexed: 08/27/2024]
Abstract
The benefit of the delivery of the right form of cancer care, tailored to the right patient, at the right time is increasingly being recognized in the global oncology community. Information on the role and feasible potential of precision oncology during the management of genitourinary cancer in Nigeria, the most populous country in Africa, is limited. This article, therefore, describes the present application of personalized medicine in Nigeria and its barriers and facilitators. It provided granular details on manpower distribution and epidemiological disparities. It also explored the use of clinical and biological markers for screening and early diagnosis, the application of team science to support genomic profiling, cost-effective approaches for image-based phenotypic precision oncology, the emerging role of molecular imaging, access to clinical trials; and their potential to support data driven diagnosis, treatment decision and care availability in order to address gaps in genitourinary cancer management in the country.
Collapse
Affiliation(s)
- Musliu Adetola Tolani
- College of Medicine, Ahmadu Bello University, Zaria, Nigeria; Division of Urology, Department of Surgery, Ahmadu Bello University Teaching Hospital, Zaria, Nigeria.
| | - Ismail Hadi Zubairu
- College of Medicine, Ahmadu Bello University, Zaria, Nigeria; Honourable Mukhtar Aliyu Betara Centre of Excellence in Oncology, Ahmadu Bello University Teaching Hospital, Zaria, Nigeria
| | - Kabir Balarabe
- College of Medicine, Ahmadu Bello University, Zaria, Nigeria; Department of Pathology, Ahmadu Bello University Teaching Hospital, Zaria, Nigeria
| | - Mudi Awaisu
- College of Medicine, Ahmadu Bello University, Zaria, Nigeria; Division of Urology, Department of Surgery, Ahmadu Bello University Teaching Hospital, Zaria, Nigeria
| | - Mubarak Abdullahi
- College of Medicine, Ahmadu Bello University, Zaria, Nigeria; Department of Radiology, Ahmadu Bello University Teaching Hospital, Zaria, Nigeria
| | | | - Shehu Salihu Umar
- College of Medicine, Ahmadu Bello University, Zaria, Nigeria; Honourable Mukhtar Aliyu Betara Centre of Excellence in Oncology, Ahmadu Bello University Teaching Hospital, Zaria, Nigeria
| | - Ahmad Bello
- College of Medicine, Ahmadu Bello University, Zaria, Nigeria; Division of Urology, Department of Surgery, Ahmadu Bello University Teaching Hospital, Zaria, Nigeria
| | - Scott T Tagawa
- Division of Hematology & Medical Oncology, Weill Cornell Medicine, New York, United States
| |
Collapse
|
152
|
Zhang Z, Zhang W, He C, Xie J, Liang F, Zhao Y, Tan L, Lai S, Jiang X, Wei X, Zhen X, Yang R. Identification of macrotrabecular-massive hepatocellular carcinoma through multiphasic CT-based representation learning method. Med Phys 2024; 51:9017-9030. [PMID: 39311438 DOI: 10.1002/mp.17401] [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: 03/18/2024] [Revised: 07/17/2024] [Accepted: 08/21/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Macrotrabecular-massive hepatocellular carcinoma (MTM-HCC) represents an aggressive subtype of HCC and is associated with poor survival. PURPOSE To investigate the performance of a representation learning-based feature fusion strategy that employs a multiphase contrast-enhanced CT (mpCECT)-based latent feature fusion (MCLFF) model for MTM-HCC identification. METHODS A total of 206 patients (54 MTM HCC, 152 non-MTM HCC) who underwent preoperative mpCECT with surgically confirmed HCC between July 2017 and December 2022 were retrospectively included from two medical centers. Multiphasic radiomics features were extracted from manually delineated volume of interest (VOI) of all lesions on each mpCECT phase. Representation learning based MCLFF model was built to fuse multiphasic features for MTM HCC prediction, and compared with competing models using other fusion methods. Conventional imaging features and clinical factors were also evaluated and analyzed. Prediction performance was validated by ROC analysis and statistical comparisons on an internal validation and an external testing dataset. RESULTS Fusion of radiomics features from the arterial phase (AP) and portal venous phase (PAP) using MCLFF demonstrated superior performance in MTM HCC prediction, with a higher AUC of 0.857 compared with all competing models in the internal validation set. Integration of multiple radiological or clinical features further improved the overall performance, with the highest AUCs of 0.857 and 0.836 respectively achieved in the internal validation and external testing set. CONCLUSIONS Multiphasic radiomics features of AP and PVP fused by the MCLFF have demonstrated substantial potential in the accurate prediction of MTM HCC. Clinical factors and Radiological features in mpCECT contribute incremental values to the developed MCLFF strategy.
Collapse
Affiliation(s)
- Zhenyang Zhang
- Department of Radiology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
- Department of Radiology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
- School of Biomedical Engineering, Southern Medical University, Guangzhou, Guangdong, China
| | - Wanli Zhang
- Department of Radiology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
- Department of Radiology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Chutong He
- Medical Imaging Center, Jinan University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Jincheng Xie
- School of Biomedical Engineering, Southern Medical University, Guangzhou, Guangdong, China
| | - Fangrong Liang
- Department of Radiology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
- Department of Radiology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yandong Zhao
- Department of Radiology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
- Department of Radiology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Lilian Tan
- Department of Radiology, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Shengsheng Lai
- School of Medical Equipment, Guangdong Food and Drug Vocational College, Guangzhou, Guangdong, China
| | - Xinqing Jiang
- Department of Radiology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
- Department of Radiology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xinhua Wei
- Department of Radiology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
- Department of Radiology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xin Zhen
- School of Biomedical Engineering, Southern Medical University, Guangzhou, Guangdong, China
- Medical Big Data Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Ruimeng Yang
- Department of Radiology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
- Department of Radiology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
153
|
Yang W, Hu P, Zuo C. Application of imaging technology for the diagnosis of malignancy in the pancreaticobiliary duodenal junction (Review). Oncol Lett 2024; 28:596. [PMID: 39430731 PMCID: PMC11487531 DOI: 10.3892/ol.2024.14729] [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: 04/15/2024] [Accepted: 09/13/2024] [Indexed: 10/22/2024] Open
Abstract
The pancreaticobiliary duodenal junction (PBDJ) is the connecting area of the pancreatic duct, bile duct and duodenum. In a broad sense, it refers to a region formed by the head of the pancreas, the pancreatic segment of the common bile duct and the intraduodenal segment, the descending and the horizontal part of the duodenum, and the soft tissue around the pancreatic head. In a narrow sense, it refers to the anatomical Vater ampulla. Due to its complex and variable anatomical features, and the diversity of pathological changes, it is challenging to make an early diagnosis of malignancy at the PBDJ and define the histological type. The unique anatomical structure of this area may be the basis for the occurrence of malignant tumors. Therefore, understanding and subclassifying the anatomical configuration of the PBDJ is of great significance for the prevention and treatment of malignant tumors at their source. The present review comprehensively discusses commonly used imaging techniques and other new technologies for diagnosing malignancy at the PBDJ, offering evidence for physicians and patients to select appropriate examination methods.
Collapse
Affiliation(s)
- Wanyi Yang
- Department of Gastroduodenal and Pancreatic Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Clinical Research Center for Tumor of Pancreaticobiliary Duodenal Junction in Hunan Province, Changsha, Hunan 410013, P.R. China
- Graduates Collaborative Training Base of Hunan Cancer Hospital, Hengyang Medical School, University of South China, Changsha, Hunan 410013, P.R. China
| | - Pingsheng Hu
- Department of Gastroduodenal and Pancreatic Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Clinical Research Center for Tumor of Pancreaticobiliary Duodenal Junction in Hunan Province, Changsha, Hunan 410013, P.R. China
| | - Chaohui Zuo
- Department of Gastroduodenal and Pancreatic Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Clinical Research Center for Tumor of Pancreaticobiliary Duodenal Junction in Hunan Province, Changsha, Hunan 410013, P.R. China
- Graduates Collaborative Training Base of Hunan Cancer Hospital, Hengyang Medical School, University of South China, Changsha, Hunan 410013, P.R. China
| |
Collapse
|
154
|
Hinzpeter R, Kulanthaivelu R, Kohan A, Murad V, Mirshahvalad SA, Avery L, Ortega C, Metser U, Hope A, Yeung J, McInnis M, Veit-Haibach P. Predictive [ 18F]-FDG PET/CT-Based Radiogenomics Modelling of Driver Gene Mutations in Non-small Cell Lung Cancer. Acad Radiol 2024; 31:5314-5323. [PMID: 38997880 DOI: 10.1016/j.acra.2024.06.038] [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: 05/07/2024] [Revised: 06/19/2024] [Accepted: 06/24/2024] [Indexed: 07/14/2024]
Abstract
RATIONALE AND OBJECTIVES To investigate whether [18F]-FDG PET/CT-derived radiomics may correlate with driver gene mutations in non-small cell lung cancer (NSCLC) patients. MATERIALS AND METHODS In this IRB-approved retrospective study, 203 patients with surgically treated NSCLC who underwent subsequent genomic analysis of the primary tumour at our institution between December 2004 and January 2014 were identified. Of those, 128 patients (mean age 62.4 ± 10.8 years; range: 35-84) received preoperative [18F]-FDG PET/CT as part of their initial staging and thus were included in the study. PET and CT image segmentation and feature extraction were performed semi-automatically with an open-source software platform (LIFEx, Version 6.30, lifexsoft.org). Molecular profiles using different next-generation sequencing (NGS) panels were collected from a web-based resource (cBioPortal.ca for Cancer genomics). Two statistical models were then built to evaluate the predictive ability of [18F]-FDG PET/CT-derived radiomics features for driver gene mutations in NSCLC. RESULTS More than half (68/128, 53%) of all tumour samples harboured three or more gene mutations. Overall, 55% of tumour samples demonstrated a mutation in TP53, 26% of samples had alterations in KRAS and 17% in EGFR. Extensive statistical analysis resulted in moderate to good predictive ability. The highest Youden Index for TP53 was achieved using combined PET/CT features (0.70), for KRAS using PET features only (0.57) and for EGFR using CT features only (0.60). CONCLUSION Our study demonstrated a moderate to good correlation between radiomics features and driver gene mutations in NSCLC, indicating increased predictive ability of genomic profiles using combined [18F]-FDG PET/CT-derived radiomics features.
Collapse
Affiliation(s)
- Ricarda Hinzpeter
- University Medical Imaging Toronto, Toronto Joint Department Medical Imaging, University Health Network, Sinai Health System, Women's College Hospital, University of Toronto, M5G 2N2, Toronto, Ontario, Canada (R.H., R.K., A.K., V.M., S.A.M., C.O., U.M., M.M., P.V.H.).
| | - Roshini Kulanthaivelu
- University Medical Imaging Toronto, Toronto Joint Department Medical Imaging, University Health Network, Sinai Health System, Women's College Hospital, University of Toronto, M5G 2N2, Toronto, Ontario, Canada (R.H., R.K., A.K., V.M., S.A.M., C.O., U.M., M.M., P.V.H.)
| | - Andres Kohan
- University Medical Imaging Toronto, Toronto Joint Department Medical Imaging, University Health Network, Sinai Health System, Women's College Hospital, University of Toronto, M5G 2N2, Toronto, Ontario, Canada (R.H., R.K., A.K., V.M., S.A.M., C.O., U.M., M.M., P.V.H.)
| | - Vanessa Murad
- University Medical Imaging Toronto, Toronto Joint Department Medical Imaging, University Health Network, Sinai Health System, Women's College Hospital, University of Toronto, M5G 2N2, Toronto, Ontario, Canada (R.H., R.K., A.K., V.M., S.A.M., C.O., U.M., M.M., P.V.H.)
| | - Seyed Ali Mirshahvalad
- University Medical Imaging Toronto, Toronto Joint Department Medical Imaging, University Health Network, Sinai Health System, Women's College Hospital, University of Toronto, M5G 2N2, Toronto, Ontario, Canada (R.H., R.K., A.K., V.M., S.A.M., C.O., U.M., M.M., P.V.H.)
| | - Lisa Avery
- Department of Biostatistics, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Canada (L.A.); Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, Canada (L.A.)
| | - Claudia Ortega
- University Medical Imaging Toronto, Toronto Joint Department Medical Imaging, University Health Network, Sinai Health System, Women's College Hospital, University of Toronto, M5G 2N2, Toronto, Ontario, Canada (R.H., R.K., A.K., V.M., S.A.M., C.O., U.M., M.M., P.V.H.)
| | - Ur Metser
- University Medical Imaging Toronto, Toronto Joint Department Medical Imaging, University Health Network, Sinai Health System, Women's College Hospital, University of Toronto, M5G 2N2, Toronto, Ontario, Canada (R.H., R.K., A.K., V.M., S.A.M., C.O., U.M., M.M., P.V.H.)
| | - Andrew Hope
- Department of Radiation Oncology, University Health Network, Toronto, Canada (A.H.)
| | - Jonathan Yeung
- Division of Thoracic Surgery, Department of Surgery, Toronto General Hospital, University Health Network, University of Toronto, Toronto, Canada (J.Y.)
| | - Micheal McInnis
- University Medical Imaging Toronto, Toronto Joint Department Medical Imaging, University Health Network, Sinai Health System, Women's College Hospital, University of Toronto, M5G 2N2, Toronto, Ontario, Canada (R.H., R.K., A.K., V.M., S.A.M., C.O., U.M., M.M., P.V.H.)
| | - Patrick Veit-Haibach
- University Medical Imaging Toronto, Toronto Joint Department Medical Imaging, University Health Network, Sinai Health System, Women's College Hospital, University of Toronto, M5G 2N2, Toronto, Ontario, Canada (R.H., R.K., A.K., V.M., S.A.M., C.O., U.M., M.M., P.V.H.)
| |
Collapse
|
155
|
Gullo RL, Brunekreef J, Marcus E, Han LK, Eskreis-Winkler S, Thakur SB, Mann R, Lipman KG, Teuwen J, Pinker K. AI Applications to Breast MRI: Today and Tomorrow. J Magn Reson Imaging 2024; 60:2290-2308. [PMID: 38581127 PMCID: PMC11452568 DOI: 10.1002/jmri.29358] [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: 12/06/2023] [Revised: 03/07/2024] [Accepted: 03/09/2024] [Indexed: 04/08/2024] Open
Abstract
In breast imaging, there is an unrelenting increase in the demand for breast imaging services, partly explained by continuous expanding imaging indications in breast diagnosis and treatment. As the human workforce providing these services is not growing at the same rate, the implementation of artificial intelligence (AI) in breast imaging has gained significant momentum to maximize workflow efficiency and increase productivity while concurrently improving diagnostic accuracy and patient outcomes. Thus far, the implementation of AI in breast imaging is at the most advanced stage with mammography and digital breast tomosynthesis techniques, followed by ultrasound, whereas the implementation of AI in breast magnetic resonance imaging (MRI) is not moving along as rapidly due to the complexity of MRI examinations and fewer available dataset. Nevertheless, there is persisting interest in AI-enhanced breast MRI applications, even as the use of and indications of breast MRI continue to expand. This review presents an overview of the basic concepts of AI imaging analysis and subsequently reviews the use cases for AI-enhanced MRI interpretation, that is, breast MRI triaging and lesion detection, lesion classification, prediction of treatment response, risk assessment, and image quality. Finally, it provides an outlook on the barriers and facilitators for the adoption of AI in breast MRI. LEVEL OF EVIDENCE: 5 TECHNICAL EFFICACY: Stage 6.
Collapse
Affiliation(s)
- Roberto Lo Gullo
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Joren Brunekreef
- AI for Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Eric Marcus
- AI for Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Lynn K Han
- Weill Cornell Medical College, New York-Presbyterian Hospital, New York, NY, USA
| | - Sarah Eskreis-Winkler
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Sunitha B Thakur
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ritse Mann
- AI for Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Kevin Groot Lipman
- AI for Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Jonas Teuwen
- AI for Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Katja Pinker
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
156
|
Xin H, Lai Q, Liu Y, Liao N, Wang Y, Liao B, Zhou K, Zhou Y, Bai Y, Chen Z, Zhou Y. Integrative radiomics analyses identify universal signature for predicting prognosis and therapeutic vulnerabilities across primary and secondary liver cancers: A multi-cohort study. Pharmacol Res 2024; 210:107535. [PMID: 39626849 DOI: 10.1016/j.phrs.2024.107535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/22/2024] [Accepted: 11/29/2024] [Indexed: 12/06/2024]
Abstract
As the hallmark of cancer, genetic and phenotypic heterogeneity leads to biomarkers that are typically tailored to specific cancer type or subtype. This specificity introduces complexities in facilitating streamlined evaluations across diverse cancer types and optimizing therapeutic outcomes. In this study, we comprehensively characterized the radiological patterns underlying liver cancer (LC) by integrating radiomics profiles from computed tomography (CT) images of hepatocellular carcinoma (HCC), intrahepatic cholangiocarcinoma (ICC), and colorectal cancer liver metastases (CRLM) through unsupervised clustering analysis. We identified three distinct radiomics clusters, displaying heterogeneity in prognosis. Subsequently, we formulated a shared prognosticator, the liver cancer radiomics signature (LCRS), by discovering and manifesting connectivity among radiomics phenotypes using GGI strategy. We validated that the LCRS is independent prognostic factor after adjusting for clinic-pathologic variables (all P < 0.05), with the LCRS-High group consistently associated with worse survival outcomes across HCC, ICC, and CRLM. However, the LCRS-High group showed clinical benefit from adjuvant chemotherapy, leading to reduced disease recurrence risk and improved survival. By contrast, the LCRS-Low group, including a subset of gastric cancer liver metastases (GCLM), exhibited more favorable response to immune checkpoint inhibitors (ICIs)-based combinational therapy (P = 0.02, hazard ratio (HR): 0.34 [95 % confidence interval (CI): 0.13-0.88]). Further analysis revealed that Notch signaling pathway was enriched in LCRS-High tumors, while LCRS-Low tumors exhibited higher infiltration of natural killer cell. These findings highlight the promise of this universal scoring model to personalize management strategies for patients with LC.
Collapse
Affiliation(s)
- Hongjie Xin
- Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qianwei Lai
- Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yanping Liu
- Department of Gastroenterology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Naying Liao
- Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ying Wang
- Department of Gastroenterology, The Fourth Hospital of Changsha, Hunan Normal University, Changsha, China
| | - Bihong Liao
- Department of Radiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Keyang Zhou
- Department of Radiology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Yuchen Zhou
- Department of General Surgery, Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yang Bai
- Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Zhihua Chen
- Department of Radiology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China.
| | - Yuanping Zhou
- Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
157
|
Xue HB, Liang ML, Xu HZ, Wang CY, Xu TW, Zhao AY. Development and validation of an individualized nomogram for predicting distant metastases in gastric cancer using a CT radiomics-clinical model. Front Oncol 2024; 14:1476340. [PMID: 39735603 PMCID: PMC11672336 DOI: 10.3389/fonc.2024.1476340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 11/12/2024] [Indexed: 12/31/2024] Open
Abstract
Purpose This study aimed to develop and validate a model for accurately assessing the risk of distant metastases in patients with gastric cancer (GC). Methods A total of 301 patients (training cohort, n = 210; testing cohort, n = 91) with GC were retrospectively collected. Relevant clinical predictors were determined through the application of univariate and multivariate logistic regression analyses. Then the clinical model was established. Venous phase computed tomography (VPCT) images were utilized to extract radiomic features, and relevant features were selected using univariate analysis, Spearman correlation coefficient, and the least absolute shrinkage and selection operator (Lasso) regression. Subsequently, radiomics scores were calculated based on the selected features. Radiomics models were constructed using five machine learning algorithms according to the screened features. Furthermore, separate joint models incorporating radiomic features and clinically independent predictors were established using traditional logistic regression algorithms and machine learning algorithms, respectively. All models were comprehensively assessed through discrimination, calibration, reclassification, and clinical benefit analysis. Results The multivariate logistic regression analysis revealed that age, histological grade, and N stage were independent predictors of distant metastases. The radiomics score was derived from 15 selected features out of a total of 944 radiomic features. The predictive performance of the joint model 1 [AUC (95% CI) 0.880 (0.811-0.949)] constructed using logistic regression is superior to that of the joint model 2 [AUC (95% CI) 0.834 (0.736-0.931)] constructed using SVM algorithm. The joint model 1 [AUC(95% CI) 0.880(0.811-0.949)], demonstrated superior performance compared to the clinical model [AUC(95% CI) 0.781(0.689-0.873)] and radiomics model [AUC(95% CI) 0.740(0.626-0.855), using LR algorithm]. The NRI and IDI values for the joint model 1 and clinical model were 0.115 (95% CI 0.014 -0.216) and 0.132 (95% CI 0.093-0.171), respectively; whereas for the joint model 1 and LR model, they were found to be 0.130 (95% CI 0.018-0.243) and 0.116 (95% CI 0.072-0.160), respectively. Decision curve analysis indicated that the joint model 1 exhibited a higher clinical net benefit than other models. Conclusions The nomogram of the joint model, integrating radiomic features and clinically independent predictors, exhibits robust predictive capability for early identification of high-risk patients with a propensity for distant metastases of GC.
Collapse
Affiliation(s)
- Hui-Bin Xue
- Department of Digestive Tumor, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Mei-Li Liang
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Huang-Zhen Xu
- Department of Digestive Tumor, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Chen-Yu Wang
- Department of Digestive Tumor, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Tian-Wen Xu
- Department of Digestive Tumor, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Ai-Yue Zhao
- Department of Oncology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| |
Collapse
|
158
|
Tang S, Yen A, Wang K, Albuquerque K, Wang J. Progression-Free Survival Prediction for Locally Advanced Cervical Cancer After Chemoradiotherapy With MRI-based Radiomics. Clin Oncol (R Coll Radiol) 2024; 38:103702. [PMID: 39706142 DOI: 10.1016/j.clon.2024.103702] [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: 02/05/2024] [Revised: 11/15/2024] [Accepted: 11/25/2024] [Indexed: 12/23/2024]
Abstract
AIMS A significant proportion of locally advanced cervical cancer (LACC) patients experience disease progression post chemoradiotherapy (CRT). Currently existing clinical variables are suboptimal predictors of treatment response. This study reported a radiomics-based model leveraging information extracted from magnetic resonance (MR) T2-weighted image (T2WI) to predict the progression-free survival (PFS) for LACC following CRT. MATERIALS AND METHODS Radiomics features were extracted from pre-treatment MR T2WI in 105 LACC patients. Following pre-feature selection and a step forward feature selection method, an optimal feature set was determined with a Cox proportional hazard (CPH) model. The PFS predictions were generated through a radiomics-clinical combined model utilized five repeated nested 5-fold cross-validation (5-fold CV). Disease progression risk was stratified into high- and low-risk groups based on the predicted PFS and assessed by Kaplan-Meier analysis. RESULTS The radiomics texture feature extracted from MR T2WI significantly predict PFS in LACC after CRT. In comparison with the model using clinical variables alone, the radiomics-clinical combined model achieves significantly improved performance in testing patient cohort, achieving higher C-index (0.748 vs 0.655) and area under the curve (0.798 vs 0.660 for 2-year PFS). Meanwhile, the proposed method significantly differentiated the high- and low-risk patients groups for disease progression (P < 0.001). CONCLUSION An MR T2WI-based radiomics and clinical combined model provided improved prognostic capabilities in predicting the PFS for LACC patients treated with CRT, outperforming a model using clinical variables alone. The incorporation of MR T2WI-based radiomics is promising in assisting in personalized management in LACC, indicating the potential of MR T2WI radiomics as imaging biomarker.
Collapse
Affiliation(s)
- S Tang
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA; Advanced Imaging and Informatics for Radiation Therapy Laboratory and Medical Artificial Intelligence and Automation Laboratory, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - A Yen
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - K Wang
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA; Advanced Imaging and Informatics for Radiation Therapy Laboratory and Medical Artificial Intelligence and Automation Laboratory, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - K Albuquerque
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - J Wang
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA; Advanced Imaging and Informatics for Radiation Therapy Laboratory and Medical Artificial Intelligence and Automation Laboratory, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
159
|
Panahi M, Habibi M, Hosseini MS. Enhancing MRI radiomics feature reproducibility and classification performance in Parkinson's disease: a harmonization approach to gray-level discretization variability. MAGMA (NEW YORK, N.Y.) 2024:10.1007/s10334-024-01215-1. [PMID: 39607667 DOI: 10.1007/s10334-024-01215-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 10/26/2024] [Accepted: 11/13/2024] [Indexed: 11/29/2024]
Abstract
OBJECTIVE This study aimed to assess the reproducibility of MRI-derived radiomic features across multiple gray-level discretization levels for classifying Parkinson's disease (PD) subtypes, and to evaluate the impact of ComBat harmonization on feature stability and machine learning performance. METHODS T1-weighted MRI scans from 140 PD patients (70 tremor-dominant, 70 postural instability gait difficulty) and 70 healthy controls were obtained from the Parkinson's progression markers initiative (PPMI) database. Radiomic features were extracted from 16 brain regions using 6 discretization levels (8, 16, 32, 64, 128, and 256 bins). ComBat harmonization was applied using a combined batch variable incorporating both scanner models and discretization levels. Intraclass correlation coefficients (ICC) and Kruskal-Wallis tests assessed feature reproducibility before and after harmonization. Support vector machine classifiers were used for PD subtype classification. RESULTS ComBat harmonization significantly improved feature reproducibility across all feature groups. The percentage of features showing excellent robustness (ICC ≥ 0.90) increased substantially after harmonization. The proportion of features significantly affected by discretization levels was reduced following harmonization. Classification accuracy improved dramatically, from a range of 0.42-0.49 before harmonization to 0.86-0.96 after harmonization across most discretization levels. AUC values similarly increased from 0.60-0.67 to 0.93-0.99 after harmonization. CONCLUSIONS ComBat harmonization significantly enhanced the reproducibility of radiomic features across discretization levels and improved PD subtype classification performance. This study highlights the importance of harmonization in radiomics research for PD and suggests potential clinical applications in personalized treatment planning.
Collapse
Affiliation(s)
- Mehdi Panahi
- Department of Computer Engineering, Payame Noor University Erbil Branch, Erbil, Iraq.
| | - Maliheh Habibi
- Department of Computer Engineering, Payame Noor University Birjand Branch, Birjand, Iran
| | | |
Collapse
|
160
|
Qi M, Lu C, Dai R, Zhang J, Hu H, Shan X. Prediction of acute pancreatitis severity based on early CT radiomics. BMC Med Imaging 2024; 24:321. [PMID: 39604925 PMCID: PMC11603661 DOI: 10.1186/s12880-024-01509-9] [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: 10/01/2024] [Accepted: 11/21/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND This study aims to develop and validate an integrated predictive model combining CT radiomics and clinical parameters for early assessment of acute pancreatitis severity. METHODS A retrospective cohort of 246 patients with acute pancreatitis was analyzed, with a 70%-30% split for training and validation groups. CT image segmentation was performed using ITK-SNAP, followed by the extraction of radiomics features. The stability of the radiomics features was assessed through inter-observer Intraclass Correlation Coefficient analysis. Feature selection was carried out using univariate analysis and least absolute shrinkage and selection operator (LASSO) regression with 10-fold cross-validation. A radiomics model was constructed through logistic regression to compute the radiomics score. Concurrently, univariate and multivariate logistic regression were employed to identify independent clinical risk factors for the clinical model. The radiomics score and clinical variables were integrated into a combined model, which was visualized with a nomogram. Model performance and net clinical benefit were evaluated through the area under the receiver operating characteristic curve (AUC), the DeLong test, and decision curve analysis. RESULTS A total of 913 radiomics features demonstrated satisfactory consistency. Eight features were selected for the radiomics model. Serum calcium, C-reactive protein, and white blood cell count were identified as independent clinical predictors. The AUC of the radiomics model was 0.871 (95% CI, 0.793-0.949) in the training cohort and 0.859 (95% CI, 0.751-0.967) in the validation cohort. The clinical model achieved AUCs of 0.833 (95% CI, 0.756-0.910) and 0.810 (95% CI, 0.692-0.929) for the training and validation cohorts, respectively. The combined model outperformed both the radiomics and clinical models, with an AUC of 0.905 (95% CI, 0.837-0.973) in the training cohort and 0.908 (95% CI, 0.824-0.992) in the validation cohort. The DeLong test confirmed superior predictive performance of the combined model over both the radiomics and clinical models in the training cohort, and over the clinical model in the validation cohort. Decision curve analysis further demonstrated that the combined model provided greater net clinical benefit than the radiomics or clinical models alone. CONCLUSION The clinical-radiomics model offers a novel tool for the early prediction of acute pancreatitis severity, providing valuable support for clinical decision-making.
Collapse
Affiliation(s)
- Mingyao Qi
- Department of Radiology, Affiliated People's Hospital of Jiangsu University, No. 8 Dianli Road, Zhenjiang, Jiangsu, P. R. China
| | - Chao Lu
- Department of Radiology, Affiliated People's Hospital of Jiangsu University, No. 8 Dianli Road, Zhenjiang, Jiangsu, P. R. China
| | - Rao Dai
- Department of Radiology, Affiliated People's Hospital of Jiangsu University, No. 8 Dianli Road, Zhenjiang, Jiangsu, P. R. China
| | - Jiulou Zhang
- Artificial Intelligence Imaging Laboratory, Nanjing Medical University, No.101 Longmian Avenue, Nanjing, Jiangsu, P. R. China
| | - Hui Hu
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, No. 155 Hanzhong Road, Nanjing, Jiangsu, P. R. China.
| | - Xiuhong Shan
- Department of Radiology, Affiliated People's Hospital of Jiangsu University, No. 8 Dianli Road, Zhenjiang, Jiangsu, P. R. China.
| |
Collapse
|
161
|
Wu XH, Ke ZB, Chen ZJ, Liu WQ, Xue YT, Chen SH, Chen DN, Zheng QS, Xue XY, Wei Y, Xu N. Periprostatic fat magnetic resonance imaging based radiomics nomogram for predicting biochemical recurrence-free survival in patients with non-metastatic prostate cancer after radical prostatectomy. BMC Cancer 2024; 24:1459. [PMID: 39604917 PMCID: PMC11600801 DOI: 10.1186/s12885-024-13207-4] [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: 02/02/2024] [Accepted: 11/15/2024] [Indexed: 11/29/2024] Open
Abstract
OBJECTIVE To build and validate a periprostatic fat magnetic resonance imaging (MRI) based radiomics nomogram for prediction of biochemical recurrence-free survival (bRFS) of patients with non-metastatic prostate cancer (PCa) receiving radical prostatectomy (RP). METHODS A retrospective study was conducted on 356 patients with non-metastatic PCa who underwent preoperative mpMRI followed by RP treatment at our institution. Radiomic features were extracted from both intratumoral region and the periprostatic fat region, which were segmented on images obtained through T2-weighted imaging (T2WI) and apparent-diffusion coefficient (ADC) imaging. Three radiomics models were developed by applying the Least absolute shrinkage and selection operator (LASSO) Cox regression, followed by Cox risk regression to construct a combined radiomics-clinical model by integrating the optimal radiomics score and clinicopathological risk factors to draw a nomogram. The predictive performance was evaluated using receiver operating characteristic (ROC) curves, Kaplan-Meier analysis and calibration curves. RESULTS One hundred and twenty-one patients (33.98%) experienced biochemical recurrence. ROC analyses showed that the Area Under the Curve (AUC) of the periprostatic fat-intratumoral radiomics model demonstrated the highest AUC at 0.921 (95%CI, 0.857-0.981), 0.875 (95%CI, 0.763-0.950), 0.854 (95%CI, 0.706-0.923) for 1-year, 3-years and 5-years bRFS. Multivariate Cox regression analysis revealed that Pathological T stage, ISUP grading group and Positive surgical margin were independent prognostic factors for predicting bRFS. A radiomics-clinical nomogram based on these clinical predictors and periprostatic fat-intratumoral radiomics score was constructed. Kaplan-Meier analyses showed that radiomics-clinical nomogram was significantly related with survival of PCa (P < 0.001); and calibration curves revealed the predicted and observed survival probability of 1-year, 3-year and 5-year bRFS had high degree of consistency in the training and validation group. The radiomics-clinical nomogram showed a significant improvement than the clinical model for 1-year (AUC, 0.944; 95%CI, 0.912-0.990 vs. AUC, 0.839; 95%CI, 0.661-0.928; P = 0.009), 3-year (AUC, 0.864; 95%CI, 0.772-0.969 vs. AUC, 0.776; 95%CI, 0.602-0.872; P = 0.008), and 5-year bRFS (AUC, 0.907; 95%CI, 0.836-0.982 vs. AUC, 0.819; 95%CI, 0.687-0.915; P = 0.027). CONCLUSIONS This study developed and validated the radiomics-clinical nomogram for the prediction of bRFS in non-metastatic PCa patients underwent RP.
Collapse
Affiliation(s)
- Xiao-Hui Wu
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, Binhai Campus of the First Affiliated Hospital, National Region Medical centre, Fujian Medical University, Fuzhou, 350212, China
| | - Zhi-Bin Ke
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, Binhai Campus of the First Affiliated Hospital, National Region Medical centre, Fujian Medical University, Fuzhou, 350212, China
| | - Ze-Jia Chen
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, Binhai Campus of the First Affiliated Hospital, National Region Medical centre, Fujian Medical University, Fuzhou, 350212, China
| | - Wen-Qi Liu
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, Binhai Campus of the First Affiliated Hospital, National Region Medical centre, Fujian Medical University, Fuzhou, 350212, China
| | - Yu-Ting Xue
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, Binhai Campus of the First Affiliated Hospital, National Region Medical centre, Fujian Medical University, Fuzhou, 350212, China
| | - Shao-Hao Chen
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, Binhai Campus of the First Affiliated Hospital, National Region Medical centre, Fujian Medical University, Fuzhou, 350212, China
| | - Dong-Ning Chen
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, Binhai Campus of the First Affiliated Hospital, National Region Medical centre, Fujian Medical University, Fuzhou, 350212, China
| | - Qing-Shui Zheng
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, Binhai Campus of the First Affiliated Hospital, National Region Medical centre, Fujian Medical University, Fuzhou, 350212, China
| | - Xue-Yi Xue
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, Binhai Campus of the First Affiliated Hospital, National Region Medical centre, Fujian Medical University, Fuzhou, 350212, China
- Fujian Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
| | - Yong Wei
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China.
- Department of Urology, Binhai Campus of the First Affiliated Hospital, National Region Medical centre, Fujian Medical University, Fuzhou, 350212, China.
| | - Ning Xu
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China.
- Department of Urology, Binhai Campus of the First Affiliated Hospital, National Region Medical centre, Fujian Medical University, Fuzhou, 350212, China.
- Fujian Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China.
| |
Collapse
|
162
|
Hong S, Hong S, Oh E, Lee WJ, Jeong WK, Kim K. Development of a flexible feature selection framework in radiomics-based prediction modeling: Assessment with four real-world datasets. Sci Rep 2024; 14:29297. [PMID: 39592859 PMCID: PMC11599926 DOI: 10.1038/s41598-024-80863-8] [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: 11/21/2024] [Indexed: 11/28/2024] Open
Abstract
There are several important challenges in radiomics research; one of them is feature selection. Since many quantitative features are non-informative, feature selection becomes essential. Feature selection methods have been mixed with filter, wrapper, and embedded methods without a rule of thumb. This study aims to develop a framework for optimal feature selection in radiomics research. We developed the framework that the optimal features were selected to quickly through controlling relevance and redundancy among features. A 'FeatureMap' was generated containing information for each step and used as a platform. Through this framework, we can explore the optimal combination of radiomics features and evaluate the predictive performance using only selected features. We assessed the framework using four real datasets. The FeatureMap generated 6 combinations, with the number of features selected varying for each combination. The predictive models obtained high performances; the highest test area under the curves (AUCs) were 0.792, 0.820, 0.846 and 0.738 in the cross-validation method, respectively. We developed a flexible framework for feature selection methods in radiomics research and assessed its usefulness using various real-world data. Our framework can assist clinicians in efficiently developing predictive models based on radiomics.
Collapse
Affiliation(s)
- Sungsoo Hong
- Department of Digital Health, Samsung Advanced Institute of Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul, Republic of Korea
| | - Sungjun Hong
- Department of Digital Health, Samsung Advanced Institute of Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul, Republic of Korea
- Medical AI Research Center, Data Science Research Institute, Research Institute for Future Medicine, Samsung Medical Center, Seoul, Republic of Korea
| | - Eunsun Oh
- Department of Radiology, Soonchunhyang University Seoul Hospital, Seoul, Republic of Korea
| | - Won Jae Lee
- Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81, Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
| | - Woo Kyoung Jeong
- Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81, Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea.
| | - Kyunga Kim
- Department of Digital Health, Samsung Advanced Institute of Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul, Republic of Korea.
- Biomedical Statistics Center, Data Science Research Institute, Research Institute for Future Medicine, Samsung Medical Center, 81, Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea.
- Department of Data Convergence & Future Medicine, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
163
|
Xu T, Zhang X, Tang H, Hua T, Xiao F, Cui Z, Tang G, Zhang L. The Value of Whole-Volume Radiomics Machine Learning Model Based on Multiparametric MRI in Predicting Triple-Negative Breast Cancer. J Comput Assist Tomogr 2024:00004728-990000000-00390. [PMID: 39631431 DOI: 10.1097/rct.0000000000001691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
OBJECTIVE This study aimed to investigate the value of radiomics analysis in the precise diagnosis of triple-negative breast cancer (TNBC) based on breast dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) and apparent diffusion coefficient (ADC) maps. METHODS This retrospective study included 326 patients with pathologically proven breast cancer (TNBC: 129, non-TNBC: 197). The lesions were segmented using the ITK-SNAP software, and whole-volume radiomics features were extracted using a radiomics platform. Radiomics features were obtained from DCE-MRI and ADC maps. The least absolute shrinkage and selection operator regression method was employed for feature selection. Three prediction models were constructed using a support vector machine classifier: Model A (based on the selected features of the ADC maps), Model B (based on the selected features of DCE-MRI), and Model C (based on the selected features of both combined). Receiver operating characteristic curves were used to evaluate the diagnostic performance of the conventional MR image model and the 3 radiomics models in predicting TNBC. RESULTS In the training dataset, the AUCs for the conventional MR image model and the 3 radiomics models were 0.749, 0.801, 0.847, and 0.896. The AUCs for the conventional MR image model and 3 radiomics models in the validation dataset were 0.693, 0.742, 0.793, and 0.876, respectively. CONCLUSIONS Radiomics based on the combination of whole volume DCE-MRI and ADC maps is a promising tool for distinguishing between TNBC and non-TNBC.
Collapse
Affiliation(s)
- Tingting Xu
- From the Department of Radiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xueli Zhang
- From the Department of Radiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Huan Tang
- Department of Radiology, Huadong Hospital of Fudan University, Shanghai, China
| | - Ting Hua
- From the Department of Radiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Fuxia Xiao
- From the Department of Radiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhijun Cui
- Department of Radiology, Chongming Branch of Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | | | - Lin Zhang
- From the Department of Radiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
164
|
Wang X, Wei M, Chen Y, Jia J, Zhang Y, Dai Y, Qin C, Bai G, Chen S. Intratumoral and peritumoral MRI-based radiomics for predicting extrapelvic peritoneal metastasis in epithelial ovarian cancer. Insights Imaging 2024; 15:281. [PMID: 39576435 PMCID: PMC11584833 DOI: 10.1186/s13244-024-01855-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 10/26/2024] [Indexed: 11/25/2024] Open
Abstract
OBJECTIVES To investigate the potential of intratumoral and peritumoral radiomics derived from T2-weighted MRI to preoperatively predict extrapelvic peritoneal metastasis (EPM) in patients with epithelial ovarian cancer (EOC). METHODS In this retrospective study, 488 patients from four centers were enrolled and divided into training (n = 245), internal test (n = 105), and external test (n = 138) sets. Intratumoral and peritumoral models were constructed based on radiomics features extracted from the corresponding regions. A combined intratumoral and peritumoral model was developed via a feature-level fusion. An ensemble model was created by integrating this combined model with specific independent clinical predictors. The robustness and generalizability of these models were assessed using tenfold cross-validation and both internal and external testing. Model performance was evaluated by the area under the receiver operating characteristic curve (AUC). The Shapley Additive Explanation method was employed for model interpretation. RESULTS The ensemble model showed superior performance across the tenfold cross-validation, with the highest mean AUC of 0.844 ± 0.063. On the internal test set, the peritumoral and ensemble models significantly outperformed the intratumoral model (AUC = 0.786 and 0.832 vs. 0.652, p = 0.007 and p < 0.001, respectively). On the external test set, the AUC of the ensemble model significantly exceeded those of the intratumoral and peritumoral models (0.843 vs. 0.750 and 0.789, p = 0.008 and 0.047, respectively). CONCLUSION Peritumoral radiomics provide more informative insights about EPM than intratumoral radiomics. The ensemble model based on MRI has the potential to preoperatively predict EPM in EOC patients. CRITICAL RELEVANCE STATEMENT Integrating both intratumoral and peritumoral radiomics information based on MRI with clinical characteristics is a promising noninvasive method to predict EPM to guide preoperative clinical decision-making for EOC patients. KEY POINTS Peritumoral radiomics can provide valuable information about extrapelvic peritoneal metastasis in epithelial ovarian cancer. The ensemble model demonstrated satisfactory performance in predicting extrapelvic peritoneal metastasis. Combining intratumoral and peritumoral MRI radiomics contributes to clinical decision-making in epithelial ovarian cancer.
Collapse
Affiliation(s)
- Xinyi Wang
- Department of Radiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| | - Mingxiang Wei
- Department of Radiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| | - Ying Chen
- Department of Radiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| | - Jianye Jia
- Department of Radiology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, China
| | - Yu Zhang
- Department of Radiology, The Fourth Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yao Dai
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Cai Qin
- Department of Radiology, Tumor Hospital Affiliated to Nantong University, Nantong, Jiangsu, China
| | - Genji Bai
- Department of Radiology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, China
| | - Shuangqing Chen
- Department of Radiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China.
| |
Collapse
|
165
|
Ding G, Li K. A CT-Based Clinical-Radiomics Nomogram for Predicting the Overall Survival to TACE Combined with Camrelizumab and Apatinib in Patients with Advanced Hepatocellular Carcinoma. Acad Radiol 2024:S1076-6332(24)00840-7. [PMID: 39578199 DOI: 10.1016/j.acra.2024.10.052] [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: 09/23/2024] [Revised: 10/16/2024] [Accepted: 10/30/2024] [Indexed: 11/24/2024]
Abstract
RATIONALE AND OBJECTIVES To construct a computed tomography (CT)-based clinical-radiomics nomogram for estimating overall survival (OS) in advanced hepatocellular carcinoma (HCC) patients receiving transcatheter arterial chemoembolization (TACE) in combination with camrelizumab and apatinib. METHODS A retrospective recruitment of 150 patients with clinically or pathologically confirmed HCC was conducted, followed by their division into training cohort (n = 105) and test cohort (n = 45). To generate the radiomics score (Rad-score), a series of analyses were performed, including Pearson correlation analysis, univariate Cox analysis, and least absolute shrinkage and selection operator Cox regression analysis. Subsequently, a clinical-radiomics nomogram was constructed using the Rad-score combined with independent clinical prognostic factors, followed by assessments of its calibration, discrimination, reclassification, and clinical utility. RESULTS Five CT radiomics features were selected. The Rad-score showed a significant correlation with OS (P < 0.001). The clinical-radiomics nomogram demonstrated superior performance in estimating OS, with a concordance index (C-index) of 0.840, compared to the radiomics nomogram (C-index: 0.817) and the clinical nomogram (C-index: 0.661). It also exhibited high 1-year and 2-year area under the curves of 0.936 and 0.946, respectively. Additionally, the clinical-radiomics nomogram markedly enhanced classification accuracy for OS outcomes, as evidenced by net reclassification improvement and integrated discrimination improvement. Decision curve analysis confirmed its clinical utility. CONCLUSION A CT-based clinical-radiomics nomogram exhibits strong potential for predicting OS in advanced HCC patients undergoing TACE combined with camrelizumab and apatinib.
Collapse
Affiliation(s)
- Guangyao Ding
- Department of General Surgery, Hefei BOE Hospital, Hefei, Anhui, China
| | - Kailang Li
- Department of General Surgery, Hefei BOE Hospital, Hefei, Anhui, China.
| |
Collapse
|
166
|
Chen M, van der Pal Z, Poirot MG, Schrantee A, Bottelier M, Kooij SJJ, Marquering HA, Reneman L, Caan MWA. Prediction of methylphenidate treatment response for ADHD using conventional and radiomics T1 and DTI features: Secondary analysis of a randomized clinical trial. Neuroimage Clin 2024; 45:103707. [PMID: 39591718 PMCID: PMC11626811 DOI: 10.1016/j.nicl.2024.103707] [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: 10/08/2024] [Revised: 11/11/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024]
Abstract
BACKGROUND Attention-Deficit/Hyperactivity Disorder (ADHD) is commonly treated with methylphenidate (MPH). Although highly effective, MPH treatment still has a relatively high non-response rate of around 30%, highlighting the need for a better understanding of treatment response. Radiomics of T1-weighted images and Diffusion Tensor Imaging (DTI) combined with machine learning approaches could offer a novel method for assessing MPH treatment response. PURPOSE To evaluate the accuracy of both conventional and radiomics approaches in predicting treatment response based on baseline T1 and DTI data in stimulant-naive ADHD participants. METHODS We performed a secondary analysis of a randomized clinical trial (ePOD-MPH), involving 47 stimulant-naive ADHD participants (23 boys aged 11.4 ± 0.4 years, 24 men aged 28.1 ± 4.3 years) who underwent 16 weeks of treatment with MPH. Baseline T1-weighted and DTI MRI scans were acquired. Treatment response was assessed at 8 weeks (during treatment) and one week after cessation of 16-week treatment (post-treatment) using the Clinical Global Impressions - Improvement scale as our primary outcome. We compared prediction accuracy using a conventional model and a radiomics model. The conventional approach included the volume of bilateral caudate, putamen, pallidum, accumbens, and hippocampus, and for DTI the mean fractional anisotropy (FA) of the entire brain white matter, bilateral Anterior Thalamic Radiation (ATR), and the splenium of the corpus callosum, totaling 14 regional features. For the radiomics approach, 380 features (shape-based and first-order statistics) were extracted from these 14 regions. XGBoost models with nested cross-validation were used and constructed for the total cohort (n = 47), as well as children (n = 23) and adults (n = 24) separately. Exact binomial tests were employed to compare model performance. RESULTS For the conventional model, balanced accuracy (bAcc) in predicting treatment response during treatment was 63 % for the total cohort, 32 % for children, and 36 % for adults (Area Under the Receiver Operating Characteristic Curve (AUC-ROC): 0.69, 0.33, 0.41 respectively). Radiomics models demonstrated bAcc's of 68 %, 64 %, and 64 % during treatment (AUC-ROCs of 0.73, 0.62, 0.69 respectively). These predictions were better than chance for both conventional and radiomics models in the total cohort (p = 0.04, p = 0.003 respectively). The radiomics models outperformed the conventional models during treatment in children only (p = 0.02). At post-treatment, performance was markedly reduced. CONCLUSION While conventional and radiomics models performed equally well in predicting clinical improvement across children and adults during treatment, radiomics features offered enhanced structural information beyond conventional region-based volume and FA averages in children. Prediction of symptom improvement one week after treatment cessation was poor, potentially due to the transient effects of stimulant treatment on symptom improvement.
Collapse
Affiliation(s)
- Mingshi Chen
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Location University of Amsterdam, Amsterdam, the Netherlands; Department of Biomedical Engineering and Physics, Amsterdam University Medical Center, Location University of Amsterdam, Amsterdam, the Netherlands.
| | - Zarah van der Pal
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Location University of Amsterdam, Amsterdam, the Netherlands
| | - Maarten G Poirot
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Location University of Amsterdam, Amsterdam, the Netherlands; Department of Biomedical Engineering and Physics, Amsterdam University Medical Center, Location University of Amsterdam, Amsterdam, the Netherlands
| | - Anouk Schrantee
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Location University of Amsterdam, Amsterdam, the Netherlands
| | - Marco Bottelier
- Child Study Center Accare, University Medical Center Groningen, Groningen, the Netherlands
| | - Sandra J J Kooij
- Department of Psychiatry, Amsterdam University Medical Center, Amsterdam, the Netherlands; Expertise Center Adult ADHD, PsyQ, The Hague, the Netherlands
| | - Henk A Marquering
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Location University of Amsterdam, Amsterdam, the Netherlands; Department of Biomedical Engineering and Physics, Amsterdam University Medical Center, Location University of Amsterdam, Amsterdam, the Netherlands
| | - Liesbeth Reneman
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Location University of Amsterdam, Amsterdam, the Netherlands
| | - Matthan W A Caan
- Department of Biomedical Engineering and Physics, Amsterdam University Medical Center, Location University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
167
|
Li Z, Fan Y, Ma J, Wang K, Li D, Zhang J, Wu Z, Wang L, Tian K. The novel developed and validated multiparametric MRI-based fusion radiomic and clinicoradiomic models predict the postoperative progression of primary skull base chordoma. Sci Rep 2024; 14:28752. [PMID: 39567620 PMCID: PMC11579367 DOI: 10.1038/s41598-024-80410-5] [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: 06/02/2024] [Accepted: 11/18/2024] [Indexed: 11/22/2024] Open
Abstract
Local progression of primary skull base chordoma (PSBC) is a sign of treatment failure. Predicting the postoperative progression of PSBC can aid in the development of individualized treatment plans to improve patients' progression-free survival (PFS) after surgery. This study aimed to develop a multiparametric MRI-based fusion radiomic model (FRM) and clinicoradiomic model (CRM) via radiomic and clinical analysis and to explore their validity in predicting postoperative progression in PSBC patients before surgery. In this retrospective study, a total of 129 patients with PSBC from our institution, including 57 patients with progression, were enrolled and randomized to the training set (TS) or the validation set (VS) at a 2:1 ratio. Radiomic features were extracted and dimensionally reduced from 3.0 T/axial T2-weighted imaging (T2WI), T1-weighted imaging (T1WI) and contrast-enhanced T1-weighted imaging (CE-T1WI) sequences for each patient, and the features were used for radiomic analysis. Univariate and multivariate Cox regression analyses were used to screen for key clinical factors. We constructed models on the basis of multivariate logistic regression analysis. Receiver operating characteristic (ROC) curve, calibration curve, and decision curve analyses were performed to evaluate the performance of the clinical model (CM), FRM and CRM. Through analysis, we found that blood supply was the only significantly different clinical factor in the CM. For the FRM, the area under the receiver operating characteristic curve (AUC) of the TS was 0.925, and the calibration curves were consistent across the TS. In the CRM, the AUC of the TS was 0.929, the calibration curve analysis was consistent for both the TS and the VS, and the DCA showed that the net benefit was greater at a threshold probability of > 0% for both the TS and the VS. Our proposed FRM can help clinicians better predict PSBC progression preoperatively, and the use of the CRM can lead to the development of more appropriate protocols to improve patients' PFS after surgery.
Collapse
Affiliation(s)
- Zekai Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Yanghua Fan
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Junpeng Ma
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Ke Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Da Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Junting Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Zhen Wu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China.
| | - Liang Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China.
| | - Kaibing Tian
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China.
| |
Collapse
|
168
|
Shen L, Huang X, Liu Y, Li Q, Bai S, Wang F, Yang Q. The value of multi-parameter radiomics combined with imaging features in predicting the therapeutic efficacy of HIFU treatment for uterine fibroids. Front Oncol 2024; 14:1499387. [PMID: 39634270 PMCID: PMC11614730 DOI: 10.3389/fonc.2024.1499387] [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: 09/20/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024] Open
Abstract
Objectives To evaluate the effectiveness of high-intensity focused ultrasound (HIFU) therapy for treating uterine fibroids by utilizing multi-sequence magnetic resonance imaging radiomic models. Methods One hundred and fifty patients in our hospital were randomly divided into a training cohort (n=120) and an internal test cohort (n=30), and forty-five patients from another hospital serving as an external test cohort. Radiomics features of uterine fibroids were extracted and selected based on preoperative T2-weighted imaging fat suppression(T2WI-FS)and contrast-enhanced T1WI(CE-T1WI)images, and logistic regression was used to develop the T2WI-FS, CE-T1WI, and combined T2WI-FS + CE-T1WI models, along with the radiomics-clinical model integrating radiomics features with imaging characteristics. The performance and clinical applicability of each model were assessed through receiver operating characteristic (ROC) curve, decision curve analysis (DCA), as well as Network Readiness Index (NRI) and Integrated Discrimination Index (IDI). Results The AUC values of the radiomics-clinical model and the T2WI-FS + CE-T1WI model were the highest. In the training cohort, the radiomics-clinical model showed higher AUC values than the T2WI-FS + CE-T1WI model, while in the internal and external testing cohorts, the AUC values of the T2WI-FS + CE-T1WI model were higher than that of the radiomics-clinical model. DCA further demonstrated that these two models achieved the greatest net benefit. NRI and IDI analyses suggested that the T2WI-FS + CE-T1WI model had higher clinical utility. Conclusions Both the T2WI-FS + CE-T1WI model and the radiomics-clinical model demonstrate higher predictive value and larger net benefit compared to other models.
Collapse
Affiliation(s)
- Li Shen
- Department of Radiology, The Affiliated Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - Xiao Huang
- Department of Radiology, The Affiliated Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - YuYao Liu
- Department of Radiology, The Affiliated Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - QingXue Li
- Department of Radiology, The Affiliated Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - ShanWei Bai
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fang Wang
- Department of Research and Development, Shanghai United Imaging Intelligent Co., Ltd, Shanghai, China
| | - Quan Yang
- Department of Radiology, The Affiliated Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
169
|
Park D. A Comprehensive Review of Performance Metrics for Computer-Aided Detection Systems. Bioengineering (Basel) 2024; 11:1165. [PMID: 39593823 PMCID: PMC11592234 DOI: 10.3390/bioengineering11111165] [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: 10/04/2024] [Revised: 11/14/2024] [Accepted: 11/18/2024] [Indexed: 11/28/2024] Open
Abstract
This paper aims to provide a structured analysis of the performance metrics used in computer-aided detection (CAD) systems, specifically focusing on lung nodule detection in computed tomography (CT) images. By examining key metrics along with their respective strengths and limitations, this study offers guidelines to assist in selecting appropriate metrics. Evaluation methods for CAD systems for lung nodule detection are primarily categorized into per-scan and per-nodule approaches. For per-scan analysis, a key metric is the area under the receiver operating characteristic (ROC) curve (AUROC), which evaluates the ability of the system to distinguish between scans with and without nodules. For per-nodule analysis, the nodule-level sensitivity at fixed false positives per scan is often used, supplemented by the free-response receiver operating characteristic (FROC) curve and the competition performance metric (CPM). However, the CPM does not provide normalized scores because it theoretically ranges from zero to infinity and largely varies depending on the characteristics of the data. To address the advantages and limitations of ROC and FROC curves, an alternative FROC (AFROC) was introduced to combine the strengths of both per-scan and per-nodule analyses. This paper discusses the principles of each metric and their relative strengths, providing insights into their clinical implications and practical utility.
Collapse
|
170
|
Zhang B, Zhu J, Xu R, Zou L, Lian Y, Xie X, Tian Y. A combined model integrating radiomics and deep learning based on multiparametric magnetic resonance imaging for classification of brain metastases. Acta Radiol 2024:2841851241292528. [PMID: 39552295 DOI: 10.1177/02841851241292528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
BACKGROUND Radiomics and deep learning (DL) can individually and efficiently identify the pathological type of brain metastases (BMs). PURPOSE To investigate the feasibility of utilizing multi-parametric MRI-based deep transfer learning radiomics (DTLR) for the classification of lung adenocarcinoma (LUAD) and non-LUAD BMs. MATERIAL AND METHODS A retrospective analysis was performed on 342 patients with 1389 BMs. These instances were randomly assigned to a training set of 273 (1179 BMs) and a testing set of 69 (210 BMs) in an 8:2 ratio. Eight machine learning algorithms were employed to construct the radiomics models. A DL model was developed using four pre-trained convolutional neural networks (CNNs). The DTLR model was formulated by integrating the optimal performing radiomics model and the DL model using a classification probability averaging approach. The area under the curve (AUC), calibration curve, and decision curve analysis (DCA) were utilized to assess the performance and clinical utility of the models. RESULTS The AUC for the optimal radiomics and DL model in the testing set were 0.824 (95% confidence interval [CI]= 0.726-0.923) and 0.775 (95% CI=0.666-0.884), respectively. The DTLR model demonstrated superior discriminatory power, achieving an AUC of 0.880 (95% CI=0.803-0.957). In addition, the DTLR model exhibited good consistency between actual and predicted probabilities based on the calibration curve and DCA analysis, indicating its significant clinical value. CONCLUSION Our study's DTLR model demonstrated high diagnostic accuracy in distinguishing LUAD from non-LUAD BMs. This method shows potential for the non-invasive identification of the histological subtype of BMs.
Collapse
Affiliation(s)
- Bo Zhang
- Department of Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, PR China
| | - Jinling Zhu
- Department of Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, PR China
| | - Ruizhe Xu
- Department of Radiation Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, PR China
| | - Li Zou
- Department of Radiation Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, PR China
| | - Yixin Lian
- Department of Respiratory & Critical Care Medicine, The Second Affiliated Hospital of Soochow University, Suzhou, PR China
| | - Xin Xie
- Department of Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, PR China
| | - Ye Tian
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, PR China
- Institute of Radiotherapy & Oncology, Soochow University, Suzhou, PR China
- Suzhou Key Laboratory for Radiation Oncology, Suzhou, PR China
| |
Collapse
|
171
|
Moro F, Ciancia M, Zace D, Vagni M, Tran HE, Giudice MT, Zoccoli SG, Mascilini F, Ciccarone F, Boldrini L, D'Antonio F, Scambia G, Testa AC. Role of artificial intelligence applied to ultrasound in gynecology oncology: A systematic review. Int J Cancer 2024; 155:1832-1845. [PMID: 38989809 DOI: 10.1002/ijc.35092] [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: 04/12/2024] [Revised: 06/03/2024] [Accepted: 06/27/2024] [Indexed: 07/12/2024]
Abstract
The aim of this paper was to explore the role of artificial intelligence (AI) applied to ultrasound imaging in gynecology oncology. Web of Science, PubMed, and Scopus databases were searched. All studies were imported to RAYYAN QCRI software. The overall quality of the included studies was assessed using QUADAS-AI tool. Fifty studies were included, of these 37/50 (74.0%) on ovarian masses or ovarian cancer, 5/50 (10.0%) on endometrial cancer, 5/50 (10.0%) on cervical cancer, and 3/50 (6.0%) on other malignancies. Most studies were at high risk of bias for subject selection (i.e., sample size, source, or scanner model were not specified; data were not derived from open-source datasets; imaging preprocessing was not performed) and index test (AI models was not externally validated) and at low risk of bias for reference standard (i.e., the reference standard correctly classified the target condition) and workflow (i.e., the time between index test and reference standard was reasonable). Most studies presented machine learning models (33/50, 66.0%) for the diagnosis and histopathological correlation of ovarian masses, while others focused on automatic segmentation, reproducibility of radiomics features, improvement of image quality, prediction of therapy resistance, progression-free survival, and genetic mutation. The current evidence supports the role of AI as a complementary clinical and research tool in diagnosis, patient stratification, and prediction of histopathological correlation in gynecological malignancies. For example, the high performance of AI models to discriminate between benign and malignant ovarian masses or to predict their specific histology can improve the diagnostic accuracy of imaging methods.
Collapse
Affiliation(s)
- Francesca Moro
- Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
| | - Marianna Ciancia
- Dipartimento Universitario Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Rome, Italy
- Dipartimento di Salute della Donna e del Bambino, Università degli studi di Padova, Padova, Italy
| | - Drieda Zace
- Infectious Disease Clinic, Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | - Marica Vagni
- Istituto di Radiologia, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Huong Elena Tran
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
| | - Maria Teresa Giudice
- Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
| | - Sofia Gambigliani Zoccoli
- Dipartimento Universitario Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Rome, Italy
- Department of Medical and Surgical Sciences for Mother, Child and Adult, University of Modena and Reggio Emilia, Azienda Ospedaliero Universitaria Policlinico, Modena, Italy
| | - Floriana Mascilini
- Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
| | - Francesca Ciccarone
- Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
| | - Luca Boldrini
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
| | | | - Giovanni Scambia
- Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
- Dipartimento Universitario Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Antonia Carla Testa
- Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
- Dipartimento Universitario Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
172
|
Liu Y, Xiao Z, Luo Y, Qiu X, Wang L, Deng J, Yang M, Lv F. Predictive value of contrast-enhanced MRI for the regrowth of residual uterine fibroids after high-intensity focused ultrasound treatment. Insights Imaging 2024; 15:274. [PMID: 39546185 PMCID: PMC11568090 DOI: 10.1186/s13244-024-01839-w] [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: 07/08/2024] [Accepted: 10/03/2024] [Indexed: 11/17/2024] Open
Abstract
OBJECTIVES To investigate whether the signal intensity (SI) ratio of residual fibroid (RF) to myometrium using Contrast-Enhanced Magnetic Resonance Imaging (CE-MRI) could predict fibroid regrowth after high-intensity focused ultrasound (HIFU) treatment. MATERIALS AND METHODS A retrospective analysis was conducted among 164 patients with uterine fibroids who underwent HIFU. To predict the RF regrowth, the SI perfusion parameters were quantified using the RF-myometrium SI ratio on CE-MRI on day 1 post-HIFU and then compared with the fibroid-myometrium SI ratio on the T2-weighted image (T2WI) and Funaki classification 1 year later. Thirty cases from another center were used as an external validation set to evaluate the performance of RF-myometrium SI ratio. RESULTS The predictive performance of the RF-myometrium SI ratio on CE-MRI on day 1 post-HIFU (Area Under Curve, AUC: 0.869) was superior to that of the preoperative and postoperative fibroid-myometrium SI ratios on the T2WI (AUC: 0.724, 0.696) and Funaki classification (AUC: 0.663, 0.623). Multivariate analysis showed that the RF- myometrium SI ratio and RF thickness were independent factors. The RF-myometrium SI ratio reflects the long-term rate of re-intervention (r = 0.455, p < 0.001). CONCLUSION The RF-myometrium SI ratio on CE-MRI exhibits greater accuracy in predicting RF regrowth compared to the SI classification and the SI ratio on T2WI. CRITICAL RELEVANCE STATEMENT The ratio of residual uterine fibroid to myometrial signal intensity on contrast-enhanced (CE)-MRI can reflect residual blood supply, predict regrowth of fibroids, and thus reflect long-term re-intervention rate and recovery situation of clinical high-intensity focused ultrasound (HIFU) treatment. KEY POINTS Contrast-enhanced (CE)-MRI can indicate the blood supply of residual uterine fibroids after high-intensity focused ultrasound (HIFU) treatment. The predictive capability of CE-MRI ratio surpasses T2WI ratio and the Funaki. Residual fibroids can serve as a measure of the long-term efficacy of HIFU.
Collapse
Affiliation(s)
- Yang Liu
- The State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Zhibo Xiao
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuanli Luo
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xueke Qiu
- The State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Lu Wang
- The State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Jinghe Deng
- The State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Mengchu Yang
- The State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Fajin Lv
- The State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China.
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
173
|
Dong Z, Cai J, Geng H, Ni B, Yuan M, Zhang Y, Xia X, Zhang H, Zhang J, Zhu C, Wai Choi U, Regmi A, Chan CI, Yan CK, Gu Y, Cao H, Zhang Z. Image-based deep learning model to predict stoma-site incisional hernia in patients with temporary ileostomy: A retrospective study. iScience 2024; 27:111235. [PMID: 39563889 PMCID: PMC11574812 DOI: 10.1016/j.isci.2024.111235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 05/22/2024] [Accepted: 10/21/2024] [Indexed: 11/21/2024] Open
Abstract
The prophylactic implantation of biological mesh can effectively prevent the occurrence of stoma-site incisional hernia (SSIH) in patients undergoing stoma retraction. Therefore, our study prospectively established and validated a mixed model, which combined radiomics, stepwise regression, and deep learning for the prediction of SSIH in patients with temporary ileostomy. The mixed model showed good discrimination of the SSIH patients on all cohorts, which outperformed deep learning, radiomics, and clinical models alone (overall area under the curve [AUC]: 0.947 in the primary cohort, 0.876 in the external validation cohort 1, and 0.776 in the external validation cohort 2). Moreover, the sensitivity, specificity, and precision for predicting SSIH were improved in the mixed model. Thus, the mixed model can provide more information for SSIH precaution and clinical decision-making.
Collapse
Affiliation(s)
- Zhongyi Dong
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, P.R. China
| | - Jianhua Cai
- Department of General Surgery, Fudan University Affiliated Huadong Hospital, Shanghai 200040, P.R. China
| | - Haigang Geng
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, P.R. China
| | - Bo Ni
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, P.R. China
| | - Mengqing Yuan
- School of Science, The Hongkong University of Science and Technology, Hongkong 999077, P.R. China
| | - Yeqian Zhang
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, P.R. China
| | - Xiang Xia
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, P.R. China
| | - Haoyu Zhang
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, P.R. China
| | - Jie Zhang
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, P.R. China
| | - Chunchao Zhu
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, P.R. China
| | - Un Wai Choi
- School of Medicine, Shanghai Jiaotong University, Shanghai 200025, P.R. China
| | - Aksara Regmi
- School of Medicine, Shanghai Jiaotong University, Shanghai 200025, P.R. China
| | - Cheok I Chan
- School of Medicine, Shanghai Jiaotong University, Shanghai 200025, P.R. China
| | - Cara Kou Yan
- School of Medicine, Shanghai Jiaotong University, Shanghai 200025, P.R. China
| | - Yan Gu
- Department of General Surgery, Fudan University Affiliated Huadong Hospital, Shanghai 200040, P.R. China
| | - Hui Cao
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, P.R. China
| | - Zizhen Zhang
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, P.R. China
| |
Collapse
|
174
|
Ma T, Wang J, Ma F, Shi J, Li Z, Cui J, Wu G, Zhao G, An Q. Visualization analysis of research hotspots and trends in MRI-based artificial intelligence in rectal cancer. Heliyon 2024; 10:e38927. [PMID: 39524896 PMCID: PMC11544045 DOI: 10.1016/j.heliyon.2024.e38927] [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: 03/02/2024] [Revised: 10/01/2024] [Accepted: 10/02/2024] [Indexed: 11/16/2024] Open
Abstract
Background Rectal cancer (RC) is one of the most common types of cancer worldwide. With the development of artificial intelligence (AI), the application of AI in preoperative evaluation and follow-up treatment of RC based on magnetic resonance imaging (MRI) has been the focus of research in this field. This review was conducted to develop comprehensive insight into the current research progress, hotspots, and future trends in AI based on MRI in RC, which remains to be studied. Methods Literature related to AI based on MRI and RC, as of November 2023, was obtained from the Web of Science Core Collection database. Visualization and bibliometric analyses of publication quantity and content were conducted to explore temporal trends, spatial distribution, collaborative networks, influential articles, keyword co-occurrence, and research directions. Results A total of 177 papers (152 original articles and 25 reviews) were identified from 24 countries/regions, 351 institutions, and 81 journals. Since 2019, the number of studies on this topic has rapidly increased. China and the United States have contributed the highest number of publications and institutions, cultivating the most intimate collaborative relationship. The highest number of articles derive from Sun Yat-sen University, while Frontiers in Oncology has published the highest number of relevant articles. Research on MRI-based AI in this field has mainly focused on preoperative diagnosis and prediction of treatment efficacy and prognosis. Conclusions This study provides an objective and comprehensive overview of the publications on MRI-based AI in RC and identifies the present research landscape, hotspots, and prospective trends in this field, which can provide valuable guidance for scholars worldwide.
Collapse
Affiliation(s)
- Tianming Ma
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Jiawen Wang
- Department of Urology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, China
| | - Fuhai Ma
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Jinxin Shi
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Zijian Li
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Jian Cui
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Guoju Wu
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Gang Zhao
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Qi An
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China
| |
Collapse
|
175
|
Chu L, Zeng D, He Y, Dong X, Li Q, Liao X, Zhao T, Chen X, Lei T, Men W, Wang Y, Wang D, Hu M, Pan Z, Tan S, Gao JH, Qin S, Tao S, Dong Q, He Y, Li S. Segregation of the regional radiomics similarity network exhibited an increase from late childhood to early adolescence: A developmental investigation. Neuroimage 2024; 302:120893. [PMID: 39426642 DOI: 10.1016/j.neuroimage.2024.120893] [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: 01/20/2024] [Revised: 09/15/2024] [Accepted: 10/17/2024] [Indexed: 10/21/2024] Open
Abstract
Brain development is characterized by an increase in structural and functional segregation, which supports the specialization of cognitive processes within the context of network neuroscience. In this study, we investigated age-related changes in morphological segregation using individual Regional Radiomics Similarity Networks (R2SNs) constructed with a longitudinal dataset of 494 T1-weighted MR scans from 309 typically developing children aged 6.2 to 13 years at baseline. Segertation indices were defined as the relative difference in connectivity strengths within and between modules and cacluated at the global, system and local levels. Linear mixed-effect models revealed longitudinal increases in both global and system segregation indices, particularly within the limbic and dorsal attention network, and decreases within the ventral attention network. Superior performance in working memory and inhibitory control was associated with higher system-level segregation indices in default, frontoparietal, ventral attention, somatomotor and subcortical systems, and lower local segregation indices in visual network regions, regardless of age. Furthermore, gene enrichment analysis revealed correlations between age-related changes in local segregation indices and regional expression levels of genes related to developmental processes. These findings provide novel insights into typical brain developmental changes using R2SN-derived segregation indices, offering a valuable tool for understanding human brain structural and cognitive maturation.
Collapse
Affiliation(s)
- Lei Chu
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science & Medical Engineering, Beihang University, Beijing 100083, China
| | - Debin Zeng
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science & Medical Engineering, Beihang University, Beijing 100083, China
| | - Yirong He
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China
| | - Xiaoxi Dong
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China
| | - Qiongling Li
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China; Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing 100875, China; IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China
| | - Xuhong Liao
- School of Systems Science, Beijing Normal University, Beijing 100875, China
| | - Tengda Zhao
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China; Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing 100875, China; IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China
| | - Xiaodan Chen
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China; Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing 100875, China; IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China
| | - Tianyuan Lei
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China; Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing 100875, China; IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China
| | - Weiwei Men
- Beijing City Key Laboratory for Medical Physics and Engineering, Institute of Heavy Ion Physics, School of Physics, Peking University, Beijing 100871, China; Zhejiang Philosophy and Social Science Laboratory for Research in Early Development and Childcare, Hangzhou Normal University, Hangzhou 311121, China
| | - Yanpei Wang
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China
| | - Daoyang Wang
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China; Zhejiang Philosophy and Social Science Laboratory for Research in Early Development and Childcare, Hangzhou Normal University, Hangzhou 311121, China
| | - Mingming Hu
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China
| | - Zhiying Pan
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China
| | - Shuping Tan
- Beijing Huilongguan Hospital, Peking University Huilongguan Clinical Medical School, Beijing 100096, China
| | - Jia-Hong Gao
- Center for MRI Research, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China; Beijing City Key Laboratory for Medical Physics and Engineering, Institute of Heavy Ion Physics, School of Physics, Peking University, Beijing 100871, China; IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Shaozheng Qin
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China; Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing 100875, China; IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China; Chinese Institute for Brain Research, Beijing 102206, China
| | - Sha Tao
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China
| | - Qi Dong
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China
| | - Yong He
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China; Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing 100875, China; IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China; Chinese Institute for Brain Research, Beijing 102206, China.
| | - Shuyu Li
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China.
| |
Collapse
|
176
|
Liu Y, Wang Y, Hu X, Wang X, Xue L, Pang Q, Zhang H, Ma Z, Deng H, Yang Z, Sun X, Men Y, Ye F, Men K, Qin J, Bi N, Zhang J, Wang Q, Hui Z. Multimodality deep learning radiomics predicts pathological response after neoadjuvant chemoradiotherapy for esophageal squamous cell carcinoma. Insights Imaging 2024; 15:277. [PMID: 39546168 PMCID: PMC11568088 DOI: 10.1186/s13244-024-01851-0] [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: 05/18/2024] [Accepted: 10/23/2024] [Indexed: 11/17/2024] Open
Abstract
OBJECTIVES This study aimed to develop and validate a deep-learning radiomics model using CT, T2, and DWI images for predicting pathological complete response (pCR) in patients with esophageal squamous cell carcinoma (ESCC) undergoing neoadjuvant chemoradiotherapy (nCRT). MATERIALS AND METHODS Patients with ESCC undergoing nCRT followed by surgery were retrospectively enrolled from three institutions and divided into training and testing cohorts. Both traditional and deep-learning radiomics features were extracted from pre-treatment CT, T2, and DWI. Multiple radiomics models were developed, both single modality and integrated, using machine learning algorithms. The models' performance was assessed using receiver operating characteristic curve analysis, with the area under the curve (AUC) as a primary metric, alongside sensitivity and specificity from the cut-off analysis. RESULTS The study involved 151 patients, among whom 63 achieved pCR. The training cohort consisted of 89 patients from Institution 1 (median age 62, 73 males) and the testing cohort included 52 patients from Institution 2 (median age 62, 41 males), and 10 in a clinical trial from Institution 3 (median age 69, 9 males). The integrated model, combining traditional and deep learning radiomics features from CT, T2, and DWI, demonstrated the best performance with an AUC of 0.868 (95% CI: 0.766-0.959), sensitivity of 88% (95% CI: 73.9-100), and specificity of 78.4% (95% CI: 63.6-90.2) in the testing cohort. This model outperformed single-modality models and the clinical model. CONCLUSION A multimodality deep learning radiomics model, utilizing CT, T2, and DWI images, was developed and validated for accurately predicting pCR of ESCC following nCRT. CRITICAL RELEVANCE STATEMENT Our research demonstrates the satisfactory predictive value of multimodality deep learning radiomics for the response of nCRT in ESCC and provides a potentially helpful tool for personalized treatment including organ preservation strategy. KEY POINTS After neoadjuvant chemoradiotherapy, patients with ESCC have pCR rates of about 40%. The multimodality deep learning radiomics model, could predict pCR after nCRT with high accuracy. The multimodality radiomics can be helpful in personalized treatment of esophageal cancer.
Collapse
Affiliation(s)
- Yunsong Liu
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yi Wang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Xinyang Hu
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Xin Wang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Liyan Xue
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qingsong Pang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Huan Zhang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Zeliang Ma
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Heping Deng
- Department of Diagnostic Radiology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Zhaoyang Yang
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xujie Sun
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yu Men
- Department of VIP Medical Services, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Feng Ye
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Kuo Men
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianjun Qin
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Nan Bi
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jing Zhang
- Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, School of Biological Science and Medical Engineering, Beihang University, Beijing, China.
| | - Qifeng Wang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China.
| | - Zhouguang Hui
- Department of VIP Medical Services, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
177
|
Hu Y, Zhang L, Qi Q, Ren S, Wang S, Yang L, Zhang J, Liu Y, Li X, Cai X, Duan S, Zhang L. Machine learning-based ultrasomics for predicting response to tyrosine kinase inhibitor in combination with anti-PD-1 antibody immunotherapy in hepatocellular carcinoma: a two-center study. Front Oncol 2024; 14:1464735. [PMID: 39610931 PMCID: PMC11602396 DOI: 10.3389/fonc.2024.1464735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 10/28/2024] [Indexed: 11/30/2024] Open
Abstract
Objective The objective of this study is to build and verify the performance of machine learning-based ultrasomics in predicting the objective response to combination therapy involving a tyrosine kinase inhibitor (TKI) and anti-PD-1 antibody for individuals with unresectable hepatocellular carcinoma (HCC). Radiomic features can reflect the internal heterogeneity of the tumor and changes in its microenvironment. These features are closely related to pathological changes observed in histology, such as cellular necrosis and fibrosis, providing crucial non-invasive biomarkers to predict patient treatment response and prognosis. Methods Clinical, pathological, and pre-treatment ultrasound image data of 134 patients with recurrent unresectable or advanced HCC who treated with a combination of TKI and anti-PD-1 antibody therapy at Henan Provincial People's Hospital and the First Affiliated Hospital of Zhengzhou University between December 2019 and November 2023 were collected and retrospectively analyzed. Using stratified random sampling, patients from the two hospitals were assigned to training cohort (n = 93) and validation cohort (n = 41) at a 7:3 ratio. After preprocessing the ultrasound images, regions of interest (ROIs) were delineated. Ultrasomic features were extracted from the images for dimensionality reduction and feature selection. By utilizing the extreme gradient boosting (XGBoost) algorithm, three models were developed: a clinical model, an ultrasomic model, and a combined model. By analyzing the area under the receiver operating characteristic (ROC) curve (AUC), specificity, sensitivity, and accuracy, the predicted performance of the models was evaluated. In addition, we identified the optimal cutoff for the radiomic score using the Youden index and applied it to stratify patients. The Kaplan-Meier (KM) survival curves were used to examine differences in progression-free survival (PFS) between the two groups. Results Twenty ultrasomic features were selected for the construction of the ultrasomic model. The AUC of the ultrasomic model for the training cohort and validation cohort were 0.999 (95%CI: 0.997-1.000) and 0.828 (95%CI: 0.690-0.966), which compared significant favorably to those of the clinical model [AUC = 0.876 (95%CI: 0.815-0.936) for the training cohort, 0.766 (95%CI: 0.597-0.935) for the validation cohort]. Compared to the ultrasomic model, the combined model demonstrated comparable performance within the training cohort (AUC = 0.977, 95%CI: 0.957-0.998) but higher performance in the validation cohort (AUC = 0.881, 95%CI: 0.758-1.000). However, there was no statistically significant difference (p > 0.05). Furthermore, ultrasomic features were associated with PFS, which was significantly different between patients with radiomic scores (Rad-score) greater than 0.057 and those with Rad-score less than 0.057 in both the training (HR = 0.488, 95% CI: 0.299-0.796, p = 0.003) and validation cohorts (HR = 0.451, 95% CI: 0.229-0.887, p = 0.02). Conclusion The ultrasomic features demonstrates excellent performance in accurately predicting the objective response to TKI in combination with anti-PD-1 antibody immunotherapy among patients with unresectable or advanced HCC.
Collapse
Affiliation(s)
- Yiwen Hu
- Department of Ultrasound, Henan University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou, China
- Henan Engineering Technology Research Center of Ultrasonic Molecular Imaging and Nanotechnology, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Linlin Zhang
- Department of Ultrasound, Henan University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou, China
- Henan Engineering Technology Research Center of Ultrasonic Molecular Imaging and Nanotechnology, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Qinghua Qi
- Department of Ultrasound, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shanshan Ren
- Department of Ultrasound, Henan University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou, China
- Henan Engineering Technology Research Center of Ultrasonic Molecular Imaging and Nanotechnology, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Simeng Wang
- Department of Ultrasound, Henan University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou, China
- Henan Engineering Technology Research Center of Ultrasonic Molecular Imaging and Nanotechnology, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Lanling Yang
- Henan Engineering Technology Research Center of Ultrasonic Molecular Imaging and Nanotechnology, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Juan Zhang
- Department of Ultrasound, Henan University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou, China
- Henan Engineering Technology Research Center of Ultrasonic Molecular Imaging and Nanotechnology, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Yuanyuan Liu
- Department of Ultrasound, Henan University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou, China
- Henan Engineering Technology Research Center of Ultrasonic Molecular Imaging and Nanotechnology, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Xiaoxiao Li
- Department of Ultrasound, Henan University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou, China
- Henan Engineering Technology Research Center of Ultrasonic Molecular Imaging and Nanotechnology, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Xiguo Cai
- Henan Rehabilitation Clinical Medical Research Center, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Shaobo Duan
- Henan Engineering Technology Research Center of Ultrasonic Molecular Imaging and Nanotechnology, Henan Provincial People’s Hospital, Zhengzhou, China
- Henan Key Laboratory of Ultrasound Imaging and Artificial Intelligence in Medicine, Henan Provincial People’s Hospital, Zhengzhou, China
- Department of Health Management, Henan University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou, China
| | - Lianzhong Zhang
- Department of Ultrasound, Henan University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou, China
- Henan Engineering Technology Research Center of Ultrasonic Molecular Imaging and Nanotechnology, Henan Provincial People’s Hospital, Zhengzhou, China
- Henan Rehabilitation Clinical Medical Research Center, Henan Provincial People’s Hospital, Zhengzhou, China
- Henan Key Laboratory of Ultrasound Imaging and Artificial Intelligence in Medicine, Henan Provincial People’s Hospital, Zhengzhou, China
| |
Collapse
|
178
|
Dai T, Gu QB, Peng YJ, Yu CL, Liu P, He YQ. Preoperative Noninvasive Prediction of Recurrence-Free Survival in Hepatocellular Carcinoma Using CT-Based Radiomics Model. J Hepatocell Carcinoma 2024; 11:2211-2222. [PMID: 39558966 PMCID: PMC11571988 DOI: 10.2147/jhc.s493044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 11/09/2024] [Indexed: 11/20/2024] Open
Abstract
Purpose This study aims to explore the value of radiomics combined with clinical parameters in predicting recurrence-free survival (RFS) after the resection of hepatocellular carcinoma (HCC). Patients and Methods In this retrospective study, a total of 322 patients with HCC who underwent contrast-enhanced computed tomography (CT) and radical surgical resection were enrolled and randomly divided into a training group (n = 223) and a validation group (n = 97). In the training group, Univariate and multivariate Cox regression analyses were employed to obtain clinical variables related to RFS for constructing the clinical model. The least absolute shrinkage and selection operator (LASSO) and multivariate Cox regression analyses were employed to construct the radiomics model, and the clinical-radiomics model was further constructed. Model prediction performance was subsequently assessed by the area under the time-dependent receiver operating characteristic curve (AUC) and calibration curve. Additionally, Kaplan-Meier analysis was used to evaluate the model's value in predicting RFS. Correlations between radiomics features and pathological parameters were analyzed. Results The clinical-radiomics model predicted RFS at 1, 2, and 3 years more accurately than the clinical or radiomics model alone (training group, AUC = 0.834, 0.765 and 0.831, respectively; validation group, AUC = 0.715, 0.710 and 0.793, respectively). The predicted high-risk subgroup based on the clinical-radiomics nomogram had shorter RFS than predicted low-risk subgroup in data sets, enabling risk stratification of various clinical subgroups. Correlation analysis revealed that the rad-score was positively related to microvascular invasion (MVI) and Edmondson-Steiner grade. Conclusion The clinical-radiomics model effectively predicts RFS in HCC patients and identifies high-risk individuals for recurrence.
Collapse
Affiliation(s)
- Ting Dai
- Department of Radiology, The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People’s Hospital), Changsha, Hunan, People’s Republic of China
| | - Qian-Biao Gu
- Department of Radiology, The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People’s Hospital), Changsha, Hunan, People’s Republic of China
| | - Ying-Jie Peng
- Department of Radiology, The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People’s Hospital), Changsha, Hunan, People’s Republic of China
| | - Chuan-Lin Yu
- Department of Radiology, The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People’s Hospital), Changsha, Hunan, People’s Republic of China
| | - Peng Liu
- Department of Radiology, The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People’s Hospital), Changsha, Hunan, People’s Republic of China
| | - Ya-Qiong He
- Department of Radiology, The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People’s Hospital), Changsha, Hunan, People’s Republic of China
| |
Collapse
|
179
|
Huang Q, Zhang P, Guo Z, Li M, Tao C, Yu Z. Comprehensive analysis of transcriptomics and radiomics revealed the potential of TEDC2 as a diagnostic marker for lung adenocarcinoma. PeerJ 2024; 12:e18310. [PMID: 39553728 PMCID: PMC11569783 DOI: 10.7717/peerj.18310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 09/24/2024] [Indexed: 11/19/2024] Open
Abstract
Background Lung adenocarcinoma (LUAD) is a widely occurring cancer with a high death rate. Radiomics, as a high-throughput method, has a wide range of applications in different aspects of the management of multiple cancers. However, the molecular mechanism of LUAD by combining transcriptomics and radiomics in order to probe LUAD remains unclear. Methods The transcriptome data and radiomics features of LUAD were extracted from the public database. Subsequently, we used weighted gene co-expression network analysis (WGCNA) and a series of machine learning algorithms including Random Forest (RF), Least Absolute Shrinkage and Selection Operator (LASSO) logistic regression, and Support Vector Machines Recursive Feature Elimination (SVM-RFE) to proceed with the screening of diagnostic genes for LUAD. In addition, the CIBERSORT and ESTIMATE algorithms were utilized to assess the association of these genes with immune profiles. The LASSO algorithm further identified the features most relevant to the expression levels of LUAD diagnostic genes and validated the model based on receiver operating characteristic (ROC), precision-recall (PR), calibration curves and decision curve analysis (DCA) curves. Finally, RT-qPCR, transwell and cell counting kit-8 (CCK8) based assays were performed to assess the expression levels and potential functions of the screened genes in LUAD cell lines. Results We screened a total of 214 modular genes with the highest correlation with LUAD samples based on WGCNA, of which 192 genes were shown to be highly expressed in LUAD patients. Subsequently, three machine learning algorithms identified a total of four genes, including UBE2T, TEDC2, RCC1, and FAM136A, as diagnostic molecules for LUAD, and the ROC curves showed that these diagnostic molecules had good diagnostic performance (AUC values of 0.989, 0.989, 989, and 0.987, respectively). The expression of these diagnostic molecules was significantly higher in tumor samples than in normal para-cancerous tissue samples and also correlated significantly and negatively with stromal and immune scores. Specifically, we also constructed a model based on TEDC2 expression consisting of seven radiomic features. Among them, the ROC and PR curves showed that the model had an AUC value of up to 0.96, respectively. Knockdown of TEDC2 slowed down the proliferation, migration and invasion efficiency of LUAD cell lines. Conclusion In this study, we screened for diagnostic markers of LUAD and developed a non-invasive radiomics model by innovatively combining transcriptomics and radiomics data. These findings contribute to our understanding of LUAD biology and offer potential avenues for further exploration in clinical practice.
Collapse
Affiliation(s)
- Qian Huang
- Department of Hepatobiliary Disease, The Third People’s Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Peng Zhang
- Radiodiagnostic Department, The 900 Hospital of the Joint Service Support Force of the People’s Liberation Army of China, Fuzhou, China
| | - Zhixu Guo
- Information Department, The 900 Hospital of the Joint Service Support Force of the People’s Liberation Army of China, Fuzhou, China
| | - Min Li
- Pathology Department, The 900 Hospital of the Joint Service Support Force of the People’s Liberation Army of China, Fuzhou, China
| | - Chao Tao
- Radiodiagnostic Department, The 900 Hospital of the Joint Service Support Force of the People’s Liberation Army of China, Fuzhou, China
| | - Zongyang Yu
- Respiratory Department, The 900 Hospital of the Joint Service Support Force of the People’s Liberation Army of China, Fuzhou, China
| |
Collapse
|
180
|
Li X, Zhang H, Hu C, Hu L, Guo H, Chen H, Li G, Huang Q, Jiang S, Zhang S, Xing Z, Wang X. Prognostic significance of collagen content in solitary fibrous tumors of the central nervous system. Front Oncol 2024; 14:1450813. [PMID: 39600632 PMCID: PMC11588704 DOI: 10.3389/fonc.2024.1450813] [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: 06/18/2024] [Accepted: 10/24/2024] [Indexed: 11/29/2024] Open
Abstract
Purpose We aimed to explore the prognostic significance of collagen content in solitary fibrous tumors (SFTs) of the central nervous system (CNS) and preliminarily investigate its relationship with magnetic resonance imaging (MRI) features of SFTs. Methods Collagen content was identified using Masson's trichrome staining, and quantitatively assessed. Radiomic methods were applied to extract quantitative MRI features of SFTs, which were then analyzed in relation to collagen content. Results The collagen content in CNS SFTs was categorized into high- and low-content groups, with a cutoff value of 6%. Survival analysis indicated a positive correlation between collagen content and overall survival (OS). In multivariate Cox regression analysis, incorporating factors such as mitosis, necrosis, Ki67, and collagen content and other indicators, collagen content emerged as an independent prognostic factor. Collagen content demonstrated a negative correlation with tumor histological phenotype, Ki67, WHO grade, mitosis, necrosis, and brain invasion. Additionally, the signal intensity of SFTs on T2-weighted imaging (T2WI) decreased with increasing collagen content. Radiomics analysis identified 1,702 features from each patient's region of interest, with 12 features showing significant differences between the high and low collagen content groups. Among the quantitative parameters and radiomic models, the combined T1- and T2WI models exhibited the highest diagnostic performance. Conclusion These findings suggest that collagen content is an independent prognostic risk factor for OS. Furthermore, combined radiomic models based on T1-and T2WI sequences may offer a more comprehensive, objective, and accurate assessment of collagen content in CNS SFTs.
Collapse
Affiliation(s)
- Xiaoling Li
- Department of Pathology, The Second Hospital of Longyan, Longyan, Fujian, China
- Department of Pathology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Department of Pathology, First Affiliated Hospital, National Regional Medical Center, Fujian Medical University, Fuzhou, Fujian, China
| | - Hua Zhang
- Department of Radiology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Department of Radiology, First Affiliated Hospital, National Regional Medical Center, Fujian Medical University, Fuzhou, Fujian, China
| | - Chengcong Hu
- Department of Pathology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Department of Pathology, First Affiliated Hospital, National Regional Medical Center, Fujian Medical University, Fuzhou, Fujian, China
| | - Liwen Hu
- Department of Pathology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Department of Pathology, First Affiliated Hospital, National Regional Medical Center, Fujian Medical University, Fuzhou, Fujian, China
| | - Huibin Guo
- Department of Pathology, The Second Hospital of Longyan, Longyan, Fujian, China
| | - Hongbao Chen
- Department of Pathology, The Second Hospital of Longyan, Longyan, Fujian, China
| | - Guoping Li
- Department of Pathology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Department of Pathology, First Affiliated Hospital, National Regional Medical Center, Fujian Medical University, Fuzhou, Fujian, China
| | - Qian Huang
- Department of Pathology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Department of Pathology, First Affiliated Hospital, National Regional Medical Center, Fujian Medical University, Fuzhou, Fujian, China
| | - Shuie Jiang
- Department of Pathology, Jianning General Hospital, Sanming, Fujian, China
| | - Sheng Zhang
- Department of Pathology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Department of Pathology, First Affiliated Hospital, National Regional Medical Center, Fujian Medical University, Fuzhou, Fujian, China
| | - Zhen Xing
- Department of Radiology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Department of Radiology, First Affiliated Hospital, National Regional Medical Center, Fujian Medical University, Fuzhou, Fujian, China
| | - Xingfu Wang
- Department of Pathology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Department of Pathology, First Affiliated Hospital, National Regional Medical Center, Fujian Medical University, Fuzhou, Fujian, China
- Department of Pathology, Jianning General Hospital, Sanming, Fujian, China
| |
Collapse
|
181
|
Yue Q, Zhang M, Jiang W, Gao L, Ye R, Hong J, Li Y. Prognostic value of FDX1, the cuprotosis key gene, and its prediction models across imaging modalities and histology. BMC Cancer 2024; 24:1381. [PMID: 39528953 PMCID: PMC11552402 DOI: 10.1186/s12885-024-13149-x] [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: 03/10/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Cuprotosis has been identified as a novel way of cell death. The key regulator ferredoxin 1 (FDX1) was explored via pan-cancer analysis, and its prediction models were proposed across seven malignancies and two imaging modalities. METHODS The prognostic value of FDX1 was explored via 1654 cases of 33 types of cancer in the Cancer Genome Atlas database. The MRI cohort of hepatocellular carcinoma in the First Affiliated Hospital of Fujian Medical University, and CT and MRI images from the Cancer Imaging Archive, REMBRANDT and Duke databases were exploited to formulate radiomic models to predict FDX1 expression. After segmentation of volumes of interest and feature extraction, the recursive feature elimination algorithm was used to screen features, logistic regression was used to model features, immunohistochemistry staining with FDX1 antibody was performed to test the radiomic model. RESULTS FDX1 was found to be prognostic in various types of cancer. The area under the receiver operating characteristic curve of radiomic models to predict FDX1 expression reached 0.825 (95% CI = 0.739-0.911). Cross-tissue compatibility was confirmed in pan-cancer validation and test cohorts. Mechanistically, the radiomic score was significantly correlated with various immunosuppressive genes and gene mutations. The radiomic score was also found to be an independent prognostic factor, making it a potentially actionable biomarker in the clinical setting. CONCLUSIONS The expression of FDX1 could be non-invasively predicted via radiomics. The radiomic patterns with biological and clinical relevance across histology and modalities could have a broad impact on a larger population of patients.
Collapse
Affiliation(s)
- Qiuyuan Yue
- Department of Radiology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, China
- Department of Radiology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou, 350004, China
| | - Mingwei Zhang
- Department of Radiotherapy, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, China
- Key Laboratory of Radiation Biology of Fujian Higher Education Institutions, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350004, China
| | - Wenying Jiang
- Department of Breast Surgery, The Third Affiliated Hospital of Soochow University, Changzhou, 213000, China
- Department of Radiology, The Third Affiliated Hospital of Soochow University, Changzhou, 213000, China
| | - Lanmei Gao
- Key Laboratory of Child Development and Learning Science of Ministry of Education, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Rongping Ye
- Department of Radiology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, China
- Department of Radiology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350212, China
| | - Jinsheng Hong
- Department of Radiotherapy, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, China.
- Key Laboratory of Radiation Biology of Fujian Higher Education Institutions, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350004, China.
| | - Yueming Li
- Department of Radiology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, China.
- Department of Radiology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350212, China.
| |
Collapse
|
182
|
Lv M, Feng Y, Zeng S, Zhang Y, Shen W, Guan W, E X, Zeng H, Zhao R, Yu J. A bibliometrics analysis based on the application of artificial intelligence in the field of radiotherapy from 2003 to 2023. Radiat Oncol 2024; 19:157. [PMID: 39529129 PMCID: PMC11552138 DOI: 10.1186/s13014-024-02551-1] [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: 04/28/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Recent research has demonstrated that the use of artificial intelligence (AI) in radiotherapy (RT) has significantly streamlined the process for physicians to treat patients with tumors; however, bibliometric studies examining the correlation between AI and RT are not available. Providing a thorough overview of the knowledge structure and research hotspots between AI and RT was the main goal of the current study. METHOD A search was conducted on the Web of Science Core Collection (WoSCC) database for publications pertaining to AI and RT between 2003 and 2023. VOSviewers, CiteSpace, and the R program "bibliometrix" were used to do the bibliometric analysis. RESULTS The analysis comprised 615 publications from 64 countries, with USA and China leading the pack. Since 2017, there have been more and more publications about RT and AI every year. The research center that made the biggest contribution to this topic was Maastricht University. The most articles published journal in this field was Frontiers in Oncology, while Medical Physics received the greatest number of citations. Dekker Andre is the author with the greatest number of published articles, while Philippe Lambin was the most often co-cited author. In the newly identified research hotspots, "autocontouring algorithm", "deep learning", and "machine learning" stand out as the main terms. CONCLUSION In fact, our bibliometric analysis offers insightful information on current research directions and advancements pertaining to the use of AI in RT. For academics looking to understand the connection between AI and RT, this study is a great resource because it highlights current research frontiers and hot trends.
Collapse
Affiliation(s)
- Minghe Lv
- Department of Radiotherapy, Shuguang Hospital, Shanghai University of Chinese Traditional Medicine, Zhang Heng Road, Pudong New Area, Shanghai, 201203, China
| | - Yue Feng
- Department of Radiotherapy, Shuguang Hospital, Shanghai University of Chinese Traditional Medicine, Zhang Heng Road, Pudong New Area, Shanghai, 201203, China
| | - Su Zeng
- Department of Radiotherapy, Shuguang Hospital, Shanghai University of Chinese Traditional Medicine, Zhang Heng Road, Pudong New Area, Shanghai, 201203, China
| | - Yang Zhang
- Department of Radiotherapy, Shuguang Hospital, Shanghai University of Chinese Traditional Medicine, Zhang Heng Road, Pudong New Area, Shanghai, 201203, China
| | - Wenhao Shen
- Department of Radiotherapy, Shuguang Hospital, Shanghai University of Chinese Traditional Medicine, Zhang Heng Road, Pudong New Area, Shanghai, 201203, China
| | - Wenhui Guan
- Department of Radiotherapy, Shuguang Hospital, Shanghai University of Chinese Traditional Medicine, Zhang Heng Road, Pudong New Area, Shanghai, 201203, China
| | - Xiangyu E
- Department of Radiotherapy, Shuguang Hospital, Shanghai University of Chinese Traditional Medicine, Zhang Heng Road, Pudong New Area, Shanghai, 201203, China
| | - Hongwei Zeng
- Department of Radiotherapy, Shuguang Hospital, Shanghai University of Chinese Traditional Medicine, Zhang Heng Road, Pudong New Area, Shanghai, 201203, China.
| | - Ruping Zhao
- Department of Radiotherapy, Shuguang Hospital, Shanghai University of Chinese Traditional Medicine, Zhang Heng Road, Pudong New Area, Shanghai, 201203, China.
| | - Jingping Yu
- Department of Radiotherapy, Changzhou Cancer Hospital, Changzhou, 213032, China.
- Department of Radiotherapy, Shuguang Hospital, Shanghai University of Chinese Traditional Medicine, Zhang Heng Road, Pudong New Area, Shanghai, 201203, China.
| |
Collapse
|
183
|
Panahi M, Hosseini MS. Impact of Harmonization on MRI Radiomics Feature Variability Across Preprocessing Methods for Parkinson's Disease Motor Subtype Classification. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2024:10.1007/s10278-024-01320-6. [PMID: 39528885 DOI: 10.1007/s10278-024-01320-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
This study aimed to assess the reproducibility of MRI-derived radiomic features across multiple preprocessing methods for classifying Parkinson's disease (PD) motor subtypes and to evaluate the impact of ComBat harmonization on feature stability and machine learning performance. T1-weighted MRI scans from 140 PD patients (70 tremor-dominant and 70 postural instability gait difficulty) and 70 healthy controls were obtained from the Parkinson's Progression Markers Initiative (PPMI) database, acquired using different scanner models. Radiomic features were extracted from 16 brain regions using various preprocessing pipelines. ComBat harmonization was applied using a combined batch variable incorporating both scanner models and preprocessing methods. Intraclass correlation coefficients (ICC) and Kruskal-Wallis tests assessed feature reproducibility before and after harmonization. Feature selection was performed using Linear Support Vector Classifier with L1 regularization. Support vector machine classifiers were used for PD subtype classification. ComBat harmonization significantly improved feature reproducibility across all feature groups. The percentage of features showing excellent robustness (ICC ≥ 0.90) increased from 40.2 to 56.3% after harmonization. First-order statistic features showed the highest robustness, with 71.11% demonstrating excellent ICC after harmonization. The proportion of features significantly affected by preprocessing methods was reduced following harmonization. Classification accuracy improved dramatically, from a range of 34-75% before harmonization to 89-96% after harmonization across all preprocessing methods. AUC values similarly increased from 0.28-0.87 to 0.95-0.99 after harmonization. ComBat harmonization significantly enhanced the reproducibility of radiomic features across preprocessing methods and improved PD motor subtype classification performance. This study highlights the importance of harmonization in radiomics research for PD and suggests potential clinical applications in personalized treatment planning.
Collapse
Affiliation(s)
- Mehdi Panahi
- Department of Computer Engineering, Payame Noor University Erbil Branch, Erbil, Iraq.
| | | |
Collapse
|
184
|
Sun X, Li S, Ma C, Fang W, Jing X, Yang C, Li H, Zhang X, Ge C, Liu B, Li Z. Glioma subtype prediction based on radiomics of tumor and peritumoral edema under automatic segmentation. Sci Rep 2024; 14:27471. [PMID: 39523433 PMCID: PMC11551193 DOI: 10.1038/s41598-024-79344-9] [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: 07/08/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024] Open
Abstract
Comprehensive and non-invasive preoperative molecular diagnosis is important for prognostic and therapy decision-making in adult-type diffuse gliomas. We employed a deep learning method for automatic segmentation of brain gliomas directly from conventional magnetic resonance imaging (MRI) scans of the tumor core and peritumoral edema regions based on available glioma MRI data provided in the BraTS2021. Three-dimensional volumes of interest were segmented from 424 cases of glioma imaging data retrospectively obtained from two medical centers using the segmentation method and radiomic features were extracted. We developed a subtype prediction model based on extracted radiomic features and analyzed significance and correlations between glioma morphological characteristics and pathological features using data from patients with adult-type diffuse glioma. The automated segmentation achieved mean Dice scores of 0.884 and 0.889 for the tumor core and whole tumor, respectively. The area under the receiver operating characteristic curve for the prediction of adult-type diffuse gliomas subtypes was 0.945. "Glioblastoma, IDH-wildtype", "Astrocytoma, IDH-mutant", and "Oligodendroglioma, IDH-mutant, 1p/19q-coded" showed AUCs of 0.96, 0.914, and 0.961, respectively, for subtype prediction. Glioma morphological characteristics, molecular and pathological levels, and clinical data showed significant differences and correlations. An automatic segmentation model for gliomas based on 3D U-Nets was developed, and the prediction model for gliomas built using the parameters obtained from the automatic segmentation model showed high overall performance.
Collapse
Affiliation(s)
- Xiangyu Sun
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, No.125 Donghu Road, WuChang, Wuhan, 430062, China
| | - Sirui Li
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuchang District, Wuhan, China
| | - Chao Ma
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, No.125 Donghu Road, WuChang, Wuhan, 430062, China
| | - Wei Fang
- Wuhan Zhongke Industrial Research Institute of Medical Science Co., Ltd., Wuhan, China
| | - Xin Jing
- Wuhan United Imaging Healthcare Surgical Technology Co., Ltd., Wuhan, China
| | - Chao Yang
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, No.125 Donghu Road, WuChang, Wuhan, 430062, China
| | - Huan Li
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuchang District, Wuhan, China
| | - Xu Zhang
- Wuhan United Imaging Healthcare Surgical Technology Co., Ltd., Wuhan, China
| | - Chuanbin Ge
- Wuhan United Imaging Healthcare Surgical Technology Co., Ltd., Wuhan, China
| | - Bo Liu
- Wuhan United Imaging Healthcare Surgical Technology Co., Ltd., Wuhan, China
| | - Zhiqiang Li
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, No.125 Donghu Road, WuChang, Wuhan, 430062, China.
| |
Collapse
|
185
|
Yu G, Zhang Z, Eresen A, Hou Q, Amirrad F, Webster S, Nauli S, Yaghmai V, Zhang Z. Predicting and Monitoring Immune Checkpoint Inhibitor Therapy Using Artificial Intelligence in Pancreatic Cancer. Int J Mol Sci 2024; 25:12038. [PMID: 39596108 PMCID: PMC11593706 DOI: 10.3390/ijms252212038] [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: 10/14/2024] [Revised: 11/01/2024] [Accepted: 11/04/2024] [Indexed: 11/28/2024] Open
Abstract
Pancreatic cancer remains one of the most lethal cancers, primarily due to its late diagnosis and limited treatment options. This review examines the challenges and potential of using immunotherapy to treat pancreatic cancer, highlighting the role of artificial intelligence (AI) as a promising tool to enhance early detection and monitor the effectiveness of these therapies. By synthesizing recent advancements and identifying gaps in the current research, this review aims to provide a comprehensive overview of how AI and immunotherapy can be integrated to develop more personalized and effective treatment strategies. The insights from this review may guide future research efforts and contribute to improving patient outcomes in pancreatic cancer management.
Collapse
Affiliation(s)
- Guangbo Yu
- Department of Biomedical Engineering, University of California, Irvine, CA 92617, USA;
| | - Zigeng Zhang
- Department of Radiological Sciences, University of California, Irvine, CA 92868, USA; (Z.Z.); (A.E.); (Q.H.); (V.Y.)
| | - Aydin Eresen
- Department of Radiological Sciences, University of California, Irvine, CA 92868, USA; (Z.Z.); (A.E.); (Q.H.); (V.Y.)
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA 92612, USA
| | - Qiaoming Hou
- Department of Radiological Sciences, University of California, Irvine, CA 92868, USA; (Z.Z.); (A.E.); (Q.H.); (V.Y.)
| | - Farideh Amirrad
- Department of Biomedical and Pharmaceutical Sciences, Harry and Diane Rinker Health Science Campus, Chapman University, Irvine, CA 92618, USA; (F.A.); (S.W.); (S.N.)
| | - Sha Webster
- Department of Biomedical and Pharmaceutical Sciences, Harry and Diane Rinker Health Science Campus, Chapman University, Irvine, CA 92618, USA; (F.A.); (S.W.); (S.N.)
| | - Surya Nauli
- Department of Biomedical and Pharmaceutical Sciences, Harry and Diane Rinker Health Science Campus, Chapman University, Irvine, CA 92618, USA; (F.A.); (S.W.); (S.N.)
- Department of Medicine, University of California Irvine, Irvine, CA 92868, USA
| | - Vahid Yaghmai
- Department of Radiological Sciences, University of California, Irvine, CA 92868, USA; (Z.Z.); (A.E.); (Q.H.); (V.Y.)
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA 92612, USA
| | - Zhuoli Zhang
- Department of Biomedical Engineering, University of California, Irvine, CA 92617, USA;
- Department of Radiological Sciences, University of California, Irvine, CA 92868, USA; (Z.Z.); (A.E.); (Q.H.); (V.Y.)
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA 92612, USA
- Department of Pathology and Laboratory Medicine, University of California Irvine, Irvine, CA 92617, USA
| |
Collapse
|
186
|
Al Mopti A, Alqahtani A, Alshehri AHD, Li C, Nabi G. Perirenal Fat CT Radiomics-Based Survival Model for Upper Tract Urothelial Carcinoma: Integrating Texture Features with Clinical Predictors. Cancers (Basel) 2024; 16:3772. [PMID: 39594727 PMCID: PMC11593147 DOI: 10.3390/cancers16223772] [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: 09/23/2024] [Revised: 10/31/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
Background: Upper tract urothelial carcinoma (UTUC) presents significant challenges in prognostication due to its rarity and complex anatomy. This study introduces a novel approach integrating perirenal fat (PRF) radiomics with clinical factors to enhance prognostic accuracy in UTUC. Methods: The study retrospectively analyzed 103 UTUC patients who underwent radical nephroureterectomy. PRF radiomics features were extracted from preoperative CT scans using a semi-automated segmentation method. Three prognostic models were developed: clinical, radiomics, and combined. Model performance was assessed using concordance index (C-index), time-dependent Area Under the Curve (AUC), and integrated Brier score. Results: The combined model demonstrated superior performance (C-index: 0.784, 95% CI: 0.707-0.861) compared to the radiomics (0.759, 95% CI: 0.678-0.840) and clinical (0.653, 95% CI: 0.547-0.759) models. Time-dependent AUC analysis revealed the radiomics model's particular strength in short-term prognosis (12-month AUC: 0.9281), while the combined model excelled in long-term predictions (60-month AUC: 0.8403). Key PRF radiomics features showed stronger prognostic value than traditional clinical factors. Conclusions: Integration of PRF radiomics with clinical data significantly improves prognostic accuracy in UTUC. This approach offers a more nuanced analysis of the tumor microenvironment, potentially capturing early signs of tumor invasion not visible through conventional imaging. The semi-automated PRF segmentation method presents advantages in reproducibility and ease of use, facilitating potential clinical implementation.
Collapse
Affiliation(s)
- Abdulrahman Al Mopti
- Centre for Medical Engineering and Technology, School of Medicine, University of Dundee, Dundee DD1 9SY, UK; (A.A.); (G.N.)
- Radiology Department, College of Applied Medical Sciences, Najran University, Najran 55461, Saudi Arabia;
| | - Abdulsalam Alqahtani
- Centre for Medical Engineering and Technology, School of Medicine, University of Dundee, Dundee DD1 9SY, UK; (A.A.); (G.N.)
- Radiology Department, College of Applied Medical Sciences, Najran University, Najran 55461, Saudi Arabia;
| | - Ali H. D. Alshehri
- Radiology Department, College of Applied Medical Sciences, Najran University, Najran 55461, Saudi Arabia;
| | - Chunhui Li
- School of Science and Engineering, University of Dundee, Dundee DD1 4HN, UK;
| | - Ghulam Nabi
- Centre for Medical Engineering and Technology, School of Medicine, University of Dundee, Dundee DD1 9SY, UK; (A.A.); (G.N.)
| |
Collapse
|
187
|
Guerrisi A, Seri E, Dolcetti V, Miseo L, Elia F, Lo Conte G, Del Gaudio G, Pacini P, Barbato A, David E, Cantisani V. A Machine Learning Model Based on Thyroid US Radiomics to Discriminate Between Benign and Malignant Nodules. Cancers (Basel) 2024; 16:3775. [PMID: 39594731 PMCID: PMC11592088 DOI: 10.3390/cancers16223775] [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: 10/07/2024] [Revised: 10/30/2024] [Accepted: 11/06/2024] [Indexed: 11/28/2024] Open
Abstract
Background/Objectives: Thyroid nodules are a very common finding, mostly benign but sometimes malignant, and thus require accurate diagnosis. Ultrasound and fine needle biopsy are the most widely used and reliable diagnostic methods to date, but they are sometimes limited in addressing benign from malignant nodules, mainly with regard to ultrasound, by the operator's experience. Radiomics, quantitative feature extraction from medical images and machine learning offer promising avenues to improve diagnosis. The aim of this work was to develop a machine learning model based on thyroid ultrasound images to classify nodules into benign and malignant classes. Methods: For this purpose, images of ultrasonography from 142 subjects were collected. Among these subjects, 40 patients (28.2%) belonged to the class "malignant" and 102 patients (71.8%) belonged to the class "benign", according to histological diagnosis from fine-needle aspiration. This image set was used for the training, cross-validation and internal testing of three different machine learning models. A robust radiomic approach was applied, under the hypothesis that the radiomic feature could capture the disease heterogeneity among the two groups. Three models consisting of four ensembles of machine learning classifiers (random forests, support vector machines and k-nearest neighbor classifiers) were developed for the binary classification task of interest. The best performing model was then externally tested on a cohort of 21 new patients. Results: The best model (ensemble of random forest) showed Receiver Operating Characteristic-Area Under the Curve (ROC-AUC) (%) of 85 (majority vote), 83.7 ** (mean) [80.2-87.2], accuracy (%) of 83, 81.2 ** [77.1-85.2], sensitivity (%) of 70, 67.5 ** [64.3-70.7], specificity (%) of 88, 86.5 ** [82-91], positive predictive value (PPV) (%) of 70, 66.5 ** [57.9-75.1] and negative predictive value (NPV) (%) of 88, 87.1 ** [85.5-88.8] (* p < 0.05, ** p < 0.005) in the internal test cohort. It achieved an accuracy of 90.5%, a sensitivity of 100%, a specificity of 86.7%, a PPV of 75% and an NPV of 100% in the external testing cohort. Conclusions: The model constituted of four ensembles of random forest classifiers could identify all the malignant nodes and the consistent majority of benign in the external testing cohort.
Collapse
Affiliation(s)
- Antonino Guerrisi
- Radiology and Diagnostic Imaging Unit, Department of Clinical and Dermatological Research, San Gallicano Dermatological Institute IRCCS, Via Elio Chianesi 53, 00144 Rome, Italy; (A.G.); (F.E.)
| | - Elena Seri
- Department of Radiological, Anatomo-Pathological Sciences, “Sapienza” University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (E.S.); (V.D.); (G.L.C.); (G.D.G.); (P.P.); (E.D.); (V.C.)
| | - Vincenzo Dolcetti
- Department of Radiological, Anatomo-Pathological Sciences, “Sapienza” University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (E.S.); (V.D.); (G.L.C.); (G.D.G.); (P.P.); (E.D.); (V.C.)
| | - Ludovica Miseo
- Radiology and Diagnostic Imaging Unit, Department of Clinical and Dermatological Research, San Gallicano Dermatological Institute IRCCS, Via Elio Chianesi 53, 00144 Rome, Italy; (A.G.); (F.E.)
| | - Fulvia Elia
- Radiology and Diagnostic Imaging Unit, Department of Clinical and Dermatological Research, San Gallicano Dermatological Institute IRCCS, Via Elio Chianesi 53, 00144 Rome, Italy; (A.G.); (F.E.)
| | - Gianmarco Lo Conte
- Department of Radiological, Anatomo-Pathological Sciences, “Sapienza” University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (E.S.); (V.D.); (G.L.C.); (G.D.G.); (P.P.); (E.D.); (V.C.)
| | - Giovanni Del Gaudio
- Department of Radiological, Anatomo-Pathological Sciences, “Sapienza” University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (E.S.); (V.D.); (G.L.C.); (G.D.G.); (P.P.); (E.D.); (V.C.)
| | - Patrizia Pacini
- Department of Radiological, Anatomo-Pathological Sciences, “Sapienza” University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (E.S.); (V.D.); (G.L.C.); (G.D.G.); (P.P.); (E.D.); (V.C.)
| | - Angelo Barbato
- Local Health Authority of Rieti, Via del Terminillo 42, 02100 Rieti, Italy;
| | - Emanuele David
- Department of Radiological, Anatomo-Pathological Sciences, “Sapienza” University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (E.S.); (V.D.); (G.L.C.); (G.D.G.); (P.P.); (E.D.); (V.C.)
- Radiology Unit 1, Department of Medical Surgical Sciences and Advanced Technologies “GF Ingrassia”, University Hospital “Policlinico G. Rodolico”, University of Catania, 95123 Catania, Italy
| | - Vito Cantisani
- Department of Radiological, Anatomo-Pathological Sciences, “Sapienza” University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (E.S.); (V.D.); (G.L.C.); (G.D.G.); (P.P.); (E.D.); (V.C.)
| |
Collapse
|
188
|
Larose M, Archambault L, Touma N, Brodeur R, Desroches F, Raymond N, Bédard-Tremblay D, LeBlanc D, Rasekh F, Hovington H, Neveu B, Vallières M, Pouliot F. Multi-task Bayesian model combining FDG-PET/CT imaging and clinical data for interpretable high-grade prostate cancer prognosis. Sci Rep 2024; 14:26928. [PMID: 39505979 PMCID: PMC11541986 DOI: 10.1038/s41598-024-77498-0] [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: 07/09/2024] [Accepted: 10/22/2024] [Indexed: 11/08/2024] Open
Abstract
We propose a fully automatic multi-task Bayesian model, named Bayesian Sequential Network (BSN), for predicting high-grade (Gleason ≥ 8) prostate cancer (PCa) prognosis using pre-prostatectomy FDG-PET/CT images and clinical data. BSN performs one classification task and five survival tasks: predicting lymph node invasion (LNI), biochemical recurrence-free survival (BCR-FS), metastasis-free survival, definitive androgen deprivation therapy-free survival, castration-resistant PCa-free survival, and PCa-specific survival (PCSS). Experiments are conducted using a dataset of 295 patients. BSN outperforms widely used nomograms on all tasks except PCSS, leveraging multi-task learning and imaging data. BSN also provides automated prostate segmentation, uncertainty quantification, personalized feature-based explanations, and introduces dynamic predictions, a novel approach that relies on short-term outcomes to refine long-term prognosis. Overall, BSN shows great promise in its ability to exploit imaging and clinicopathological data to predict poor outcome patients that need treatment intensification with loco-regional or systemic adjuvant therapy for high-risk PCa.
Collapse
Affiliation(s)
- Maxence Larose
- Département de physique, de génie physique et d'optique, et Centre de recherche sur le cancer, Université Laval, Québec, QC, Canada.
- CHU de Québec - Université Laval et CRCHU de Québec, Québec, QC, Canada.
| | - Louis Archambault
- Département de physique, de génie physique et d'optique, et Centre de recherche sur le cancer, Université Laval, Québec, QC, Canada.
- CHU de Québec - Université Laval et CRCHU de Québec, Québec, QC, Canada.
| | - Nawar Touma
- CHU de Québec - Université Laval et CRCHU de Québec, Québec, QC, Canada
| | - Raphaël Brodeur
- Département de physique, de génie physique et d'optique, et Centre de recherche sur le cancer, Université Laval, Québec, QC, Canada
- CHU de Québec - Université Laval et CRCHU de Québec, Québec, QC, Canada
| | - Félix Desroches
- Département de physique, de génie physique et d'optique, et Centre de recherche sur le cancer, Université Laval, Québec, QC, Canada
- CHU de Québec - Université Laval et CRCHU de Québec, Québec, QC, Canada
| | - Nicolas Raymond
- Department of Computer Science, Université de Sherbrooke, Sherbrooke, QC, Canada
| | | | - Danahé LeBlanc
- Département de physique, de génie physique et d'optique, et Centre de recherche sur le cancer, Université Laval, Québec, QC, Canada
- CHU de Québec - Université Laval et CRCHU de Québec, Québec, QC, Canada
| | - Fatemeh Rasekh
- CHU de Québec - Université Laval et CRCHU de Québec, Québec, QC, Canada
| | - Hélène Hovington
- CHU de Québec - Université Laval et CRCHU de Québec, Québec, QC, Canada
| | - Bertrand Neveu
- CHU de Québec - Université Laval et CRCHU de Québec, Québec, QC, Canada
| | - Martin Vallières
- Department of Computer Science, Université de Sherbrooke, Sherbrooke, QC, Canada.
| | - Frédéric Pouliot
- CHU de Québec - Université Laval et CRCHU de Québec, Québec, QC, Canada.
| |
Collapse
|
189
|
Koçak B, D’Antonoli TA, Cuocolo R. Exploring radiomics research quality scoring tools: a comparative analysis of METRICS and RQS. Diagn Interv Radiol 2024; 30:366-369. [PMID: 38700426 PMCID: PMC11589524 DOI: 10.4274/dir.2024.242793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 04/25/2024] [Indexed: 05/05/2024]
Affiliation(s)
- Burak Koçak
- University of Health Sciences, Başakşehir Çam and Sakura City Hospital, Clinic of Radiology, İstanbul, Türkiye
| | - Tugba Akinci D’Antonoli
- Cantonal Hospital Baselland, Institute of Radiology and Nuclear Medicine, Liestal, Switzerland
| | - Renato Cuocolo
- University of Salerno, Department of Medicine, Surgery and Dentistry, Baronissi, Italy
| |
Collapse
|
190
|
Wang S, Belemlilga D, Lei Y, Ganti AKP, Lin C, Asif S, Marasco JT, Oh K, Zhou S. Enhancing Survival Outcome Predictions in Metastatic Non-Small Cell Lung Cancer Through PET Radiomics Analysis. Cancers (Basel) 2024; 16:3731. [PMID: 39594686 PMCID: PMC11592397 DOI: 10.3390/cancers16223731] [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: 09/26/2024] [Revised: 10/26/2024] [Accepted: 10/30/2024] [Indexed: 11/28/2024] Open
Abstract
(1) Background: Advanced-stage lung cancer poses significant management challenges. The goal of this study was to identify crucial clinical and PET radiomics features that enable prognostic stratification for predicting outcomes. (2) Methods: PET radiomics features of the primary lung lesions were extracted from 99 patients with stage IVB NSCLC, and the robustness of these PET radiomics features was evaluated against uncertainties stemming from extraction parameters and contour variation. We trained three survival risk models (clinical, radiomics, and a composite) through a penalized Cox model framework. We also created a Balanced Random Forest classification predictive model, using the selected features, to predict 1-year survival. (3) Results: We identified 367 common PET radiomics features that exhibited robustness to perturbations introduced by contour variation and extraction parameters. Our findings indicated that both the radiomics and the composite model outperformed the clinical model in stratifying the risk for survival with statistical significance. In predicting 1-year survival, the radiomics model and the composite model also achieved better predicting accuracies compared to the clinical model. (4) Conclusions: Robust PET radiomics analysis successfully facilitated the stratification of patient risk for survival outcomes and predicted 1-year survival in stage IVB NSCLC.
Collapse
Affiliation(s)
- Shuo Wang
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (D.B.); (Y.L.); (C.L.); (J.T.M.); (K.O.); (S.Z.)
| | - Darryl Belemlilga
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (D.B.); (Y.L.); (C.L.); (J.T.M.); (K.O.); (S.Z.)
- College of Arts and Sciences, University of Nebraska Omaha, Omaha, NE 68182, USA
| | - Yu Lei
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (D.B.); (Y.L.); (C.L.); (J.T.M.); (K.O.); (S.Z.)
| | - Apar Kishor P Ganti
- Division of Oncology and Hematology, Department of Internal Medicine, VA Nebraska Western Iowa Health Care System, Omaha, NE 68105, USA;
- Division of Oncology and Hematology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68105, USA;
| | - Chi Lin
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (D.B.); (Y.L.); (C.L.); (J.T.M.); (K.O.); (S.Z.)
| | - Samia Asif
- Division of Oncology and Hematology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68105, USA;
| | - Jacob T Marasco
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (D.B.); (Y.L.); (C.L.); (J.T.M.); (K.O.); (S.Z.)
| | - Kyuhak Oh
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (D.B.); (Y.L.); (C.L.); (J.T.M.); (K.O.); (S.Z.)
| | - Sumin Zhou
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (D.B.); (Y.L.); (C.L.); (J.T.M.); (K.O.); (S.Z.)
| |
Collapse
|
191
|
Song R, Liu B, Xu H. CT-based deep learning model for predicting the success of extracorporeal shock wave lithotripsy in treating ureteral stones larger than 1 cm. Urolithiasis 2024; 52:157. [PMID: 39499273 DOI: 10.1007/s00240-024-01656-2] [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: 09/23/2024] [Accepted: 10/29/2024] [Indexed: 11/07/2024]
Abstract
OBJECTIVES To develop a deep learning (DL) model based on computed tomography (CT) images to predict the success of extracorporeal shock wave lithotripsy (SWL) treatment for patients with ureteral stones larger than 1 cm. MATERIALS AND METHODS We enrolled 333 patients who underwent SWL treatment for ureteral stones and randomly divided them into training and test sets. A DL model was built based on CT images of ureteral stones to predict SWL outcomes. The predictive efficacy of the DL model was assessed by comparing it with traditional and radiomics models. RESULTS The DL model demonstrated significantly better predictive performance in both training and test sets compared to radiomics (training set, AUC: 0.993 vs. 0.923, P < 0.001; test set AUC: 0.982 vs. 0.846, P < 0.001) and traditional models (training set AUC: 0.993 vs. 0.75, P = 0.005; test set AUC: 0.982 vs. 0.677, P < 0.001). Decision curve analysis (DCA) also proved that the DL model brought more benefit in predicting the success of SWL treatment than other methods. CONCLUSION The DL model based on CT images showed excellent ability to predict the probability of success of SWL treatment for patients with ureteral stones larger than 1 cm, providing a new auxiliary tool for clinical treatment decision-making.
Collapse
Affiliation(s)
- Rijin Song
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, Jiangsu Province, China
| | - Bo Liu
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, Jiangsu Province, China
| | - Huixin Xu
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, Jiangsu Province, China.
| |
Collapse
|
192
|
Cè M, Chiriac MD, Cozzi A, Macrì L, Rabaiotti FL, Irmici G, Fazzini D, Carrafiello G, Cellina M. Decoding Radiomics: A Step-by-Step Guide to Machine Learning Workflow in Hand-Crafted and Deep Learning Radiomics Studies. Diagnostics (Basel) 2024; 14:2473. [PMID: 39594139 PMCID: PMC11593328 DOI: 10.3390/diagnostics14222473] [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: 09/04/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024] Open
Abstract
Although radiomics research has experienced rapid growth in recent years, with numerous studies dedicated to the automated extraction of diagnostic and prognostic information from various imaging modalities, such as CT, PET, and MRI, only a small fraction of these findings has successfully transitioned into clinical practice. This gap is primarily due to the significant methodological challenges involved in radiomics research, which emphasize the need for a rigorous evaluation of study quality. While many technical aspects may lie outside the expertise of most radiologists, having a foundational knowledge is essential for evaluating the quality of radiomics workflows and contributing, together with data scientists, to the development of models with a real-world clinical impact. This review is designed for the new generation of radiologists, who may not have specialized training in machine learning or radiomics, but will inevitably play a role in this evolving field. The paper has two primary objectives: first, to provide a clear, systematic guide to radiomics study pipeline, including study design, image preprocessing, feature selection, model training and validation, and performance evaluation. Furthermore, given the critical importance of evaluating the robustness of radiomics studies, this review offers a step-by-step guide to the application of the METhodological RadiomICs Score (METRICS, 2024)-a newly proposed tool for assessing the quality of radiomics studies. This roadmap aims to support researchers and reviewers alike, regardless of their machine learning expertise, in utilizing this tool for effective study evaluation.
Collapse
Affiliation(s)
- Maurizio Cè
- Postgraduation School in Radiodiagnostics, Università degli Studi di Milano, Via Festa del Perdono 7, 20122 Milan, Italy
| | | | - Andrea Cozzi
- Imaging Institute of Southern Switzerland (IIMSI), Ente Ospedaliero Cantonale (EOC), Via Tesserete 46, 6900 Lugano, Switzerland;
| | - Laura Macrì
- Postgraduation School in Radiodiagnostics, Università degli Studi di Milano, Via Festa del Perdono 7, 20122 Milan, Italy
| | - Francesca Lucrezia Rabaiotti
- Postgraduation School in Radiodiagnostics, Università degli Studi di Milano, Via Festa del Perdono 7, 20122 Milan, Italy
| | - Giovanni Irmici
- Breast Imaging Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Giacomo Venezian 1, 20133 Milan, Italy
| | - Deborah Fazzini
- Radiology Department, Centro Diagnostico Italiano, Via Saint Bon 20, 20147 Milan, Italy
| | - Gianpaolo Carrafiello
- Radiology Department, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122 Milan, Italy
- Department of Oncology and Hematology-Oncology, Università degli Studi di Milano, Via Festa del Perdono 7, 20122 Milan, Italy
| | - Michaela Cellina
- Radiology Department, ASST Fatebenefratelli Sacco, Piazza Principessa Clotilde 3, 20121 Milan, Italy
| |
Collapse
|
193
|
Mylona E, Zaridis DI, Kalantzopoulos CΝ, Tachos NS, Regge D, Papanikolaou N, Tsiknakis M, Marias K, Fotiadis DI. Optimizing radiomics for prostate cancer diagnosis: feature selection strategies, machine learning classifiers, and MRI sequences. Insights Imaging 2024; 15:265. [PMID: 39495422 PMCID: PMC11535140 DOI: 10.1186/s13244-024-01783-9] [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: 03/21/2024] [Accepted: 06/27/2024] [Indexed: 11/05/2024] Open
Abstract
OBJECTIVES Radiomics-based analyses encompass multiple steps, leading to ambiguity regarding the optimal approaches for enhancing model performance. This study compares the effect of several feature selection methods, machine learning (ML) classifiers, and sources of radiomic features, on models' performance for the diagnosis of clinically significant prostate cancer (csPCa) from bi-parametric MRI. METHODS Two multi-centric datasets, with 465 and 204 patients each, were used to extract 1246 radiomic features per patient and MRI sequence. Ten feature selection methods, such as Boruta, mRMRe, ReliefF, recursive feature elimination (RFE), random forest (RF) variable importance, L1-lasso, etc., four ML classifiers, namely SVM, RF, LASSO, and boosted generalized linear model (GLM), and three sets of radiomics features, derived from T2w images, ADC maps, and their combination, were used to develop predictive models of csPCa. Their performance was evaluated in a nested cross-validation and externally, using seven performance metrics. RESULTS In total, 480 models were developed. In nested cross-validation, the best model combined Boruta with Boosted GLM (AUC = 0.71, F1 = 0.76). In external validation, the best model combined L1-lasso with boosted GLM (AUC = 0.71, F1 = 0.47). Overall, Boruta, RFE, L1-lasso, and RF variable importance were the top-performing feature selection methods, while the choice of ML classifier didn't significantly affect the results. The ADC-derived features showed the highest discriminatory power with T2w-derived features being less informative, while their combination did not lead to improved performance. CONCLUSION The choice of feature selection method and the source of radiomic features have a profound effect on the models' performance for csPCa diagnosis. CRITICAL RELEVANCE STATEMENT This work may guide future radiomic research, paving the way for the development of more effective and reliable radiomic models; not only for advancing prostate cancer diagnostic strategies, but also for informing broader applications of radiomics in different medical contexts. KEY POINTS Radiomics is a growing field that can still be optimized. Feature selection method impacts radiomics models' performance more than ML algorithms. Best feature selection methods: RFE, LASSO, RF, and Boruta. ADC-derived radiomic features yield more robust models compared to T2w-derived radiomic features.
Collapse
Affiliation(s)
- Eugenia Mylona
- Biomedical Research Institute, FORTH, GR 45110, Ioannina, Greece
- Unit of Medical Technology Intelligent Information Systems, University of Ioannina, Ioannina, Greece
| | - Dimitrios I Zaridis
- Biomedical Research Institute, FORTH, GR 45110, Ioannina, Greece
- Unit of Medical Technology Intelligent Information Systems, University of Ioannina, Ioannina, Greece
- Biomedical Engineering Laboratory, School of Electrical & Computer Engineering, National Technical University of Athens, Athens, Greece
| | - Charalampos Ν Kalantzopoulos
- Biomedical Research Institute, FORTH, GR 45110, Ioannina, Greece
- Unit of Medical Technology Intelligent Information Systems, University of Ioannina, Ioannina, Greece
| | - Nikolaos S Tachos
- Biomedical Research Institute, FORTH, GR 45110, Ioannina, Greece
- Unit of Medical Technology Intelligent Information Systems, University of Ioannina, Ioannina, Greece
| | - Daniele Regge
- Department of Radiology, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | | | - Manolis Tsiknakis
- Computational Biomedicine Laboratory, Institute of Computer Science, FORTH, GR 70013, Heraklion, Greece
- Department of Electrical and Computer Engineering, Hellenic Mediterranean University, GR 71004, Heraklion, Greece
| | - Kostas Marias
- Computational Biomedicine Laboratory, Institute of Computer Science, FORTH, GR 70013, Heraklion, Greece
- Department of Electrical and Computer Engineering, Hellenic Mediterranean University, GR 71004, Heraklion, Greece
| | - Dimitrios I Fotiadis
- Biomedical Research Institute, FORTH, GR 45110, Ioannina, Greece.
- Unit of Medical Technology Intelligent Information Systems, University of Ioannina, Ioannina, Greece.
| |
Collapse
|
194
|
Gao Z, Dai Z, Ouyang Z, Li D, Tang S, Li P, Liu X, Jiang Y, Song D. Radiomics analysis in differentiating osteosarcoma and chondrosarcoma based on T2-weighted imaging and contrast-enhanced T1-weighted imaging. Sci Rep 2024; 14:26594. [PMID: 39496777 PMCID: PMC11535035 DOI: 10.1038/s41598-024-78245-1] [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: 09/21/2023] [Accepted: 10/29/2024] [Indexed: 11/06/2024] Open
Abstract
This study was performed to investigate the diagnostic value of radiomics models constructed by fat suppressed T2-weighted imaging (T2WI-FS) and contrast-enhanced T1-weighted imaging (CET1) based on magnetic resonance imaging (MRI) for differentiation of osteosarcoma (OS) and chondrosarcoma (CS). In this retrospective cohort study, we included all inpatients with pathologically confirmed OS or CS from Second Xiangya Hospital of Central South University (Hunan, China) as of October 2020. Demographic and imaging variables were extracted from electronic medical records and compared between OS and CS group. Totals of 530 radiomics features were extracted from CET1 and T2WI-FS sequences based on MRI. The least absolute shrinkage and selection operator (LASSO) method was used for screening and dimensionality reduction of the radiomics model. Multivariate logistic regression analysis was performed to construct the radiomics model, and receiver operating characteristic curve (ROC) was generated to evaluate the diagnostic accuracy of the radiomics model. The training cohort and validation cohort included 87 and 29 patients, respectively. 8 CET1 features and 15 T2WI-FS features were screened based on the radiomics features. In the training group, the area under the receiver-operator characteristic curve (AUC) value for CET1 and T2WI-FS sequences in the radiomics model was 0.894 (95% CI 0.817-0.970) and 0.970 (95% CI 0.940-0.999), respectively. In the validation group, the AUC value for CET1 and T2WI-FS sequences in the radiomics model was 0.821 (95% CI 0.642-1.000) and 0.899 (95% CI 0.785-1.000), respectively. In this study, we developed a radiomics model based on T2WI-FS and CET1 sequences to differentiate between OS and CS. This model exhibits good performance and can help clinicians make decisions and optimize the use of healthcare resources.
Collapse
Affiliation(s)
- Zhi Gao
- Department of Infectious Diseases, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, People's Republic of China
- FuRong Laboratory, Changsha, 410078, Hunan, China
- Clinical Research Center for Viral Hepatitis in Hunan Province, Changsha, Hunan, China
| | - Zhongshang Dai
- Department of Infectious Diseases, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, People's Republic of China
- FuRong Laboratory, Changsha, 410078, Hunan, China
- Clinical Research Center for Viral Hepatitis in Hunan Province, Changsha, Hunan, China
| | - Zhengxiao Ouyang
- Department of Orthopedics, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, People's Republic of China
| | - Dianqing Li
- The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Sihuai Tang
- Department of Orthopedics, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, People's Republic of China
| | - Penglin Li
- Department of Orthopedics, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, People's Republic of China
| | - Xudong Liu
- Department of Orthopedics, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, People's Republic of China
| | - Yongfang Jiang
- Department of Infectious Diseases, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, People's Republic of China.
- FuRong Laboratory, Changsha, 410078, Hunan, China.
- Clinical Research Center for Viral Hepatitis in Hunan Province, Changsha, Hunan, China.
| | - Deye Song
- Department of Orthopedics, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, People's Republic of China.
| |
Collapse
|
195
|
Zhang H, Teng C, Yao Y, Bian W, Chen J, Liu H, Wang Z. MRI-based radiomics models for noninvasive evaluation of lymphovascular space invasion in cervical cancer: a systematic review and meta-analysis. Clin Radiol 2024; 79:e1372-e1382. [PMID: 39183137 DOI: 10.1016/j.crad.2024.07.018] [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: 04/24/2024] [Revised: 07/02/2024] [Accepted: 07/26/2024] [Indexed: 08/27/2024]
Abstract
AIM Aimed to evaluate the diagnostic performance of preoperative MRI-based radiomic models for noninvasive prediction of lymphovascular space invasion (LVSI) in patients with cervical cancer (CC). MATERIALS AND METHODS A systematic search of the PubMed, Embase, Web of Science, and Cochrane databases was conducted up to December 21, 2023. The quality of the studies was assessed utilizing the Radiomics Quality Score (RQS) system and the Quality Assessment of Diagnostic Accuracy Studies (QUADAS-2) tool. Pooled estimates of sensitivity, specificity, diagnostic odds ratio (DOR), and area under the curve (AUC) of the summary receiver operating characteristic curve (SROC) were computed. The clinical utility was evaluated using the Fagan nomogram. Heterogeneity was investigated and subgroup analyses were conducted. RESULTS Eleven studies were included, with nine studies reporting independent validation sets. In the training sets, the pooled sensitivity, specificity, DOR, and AUC of SROC were 0.81 (95% CI: 0.75-0.85), 0.78 (95% CI: 0.73-0.83), 15 (95% CI: 11-20), and 0.86 (95% CI: 0.79-0.92), respectively. For the validation sets, the overall sensitivity, specificity, DOR, and AUC of SROC were 0.79 (95% CI: 0.73-0.84), 0.73 (95% CI: 0.67-0.78), 10 (95% CI: 7-15), and 0.83 (95% CI: 0.71-0.91), respectively. The Fagan nomogram indicated good clinical utility. Subgroup analysis revealed that multi-sequence MRI-based models exhibited superior diagnostic performance compared to single-sequence MRI-based models in validation sets. CONCLUSION This meta-analysis highlights the potential diagnostic efficacy of MRI-based radiomic models for predicting LVSI in CC. Nevertheless, large-sample, multicenter studies are still warranted, and improvements in the standardization of radiomics methodology are necessary.
Collapse
Affiliation(s)
- H Zhang
- Department of Radiology, Jiaxing Maternity and Child Health Care Hospital, Jiaxing, Zhejiang 314000, China
| | - C Teng
- Department of Radiology, Wenzhou Central Hospital, Wenzhou, Zhejiang 325000, China
| | - Y Yao
- Department of Radiology, Jiaxing Maternity and Child Health Care Hospital, Jiaxing, Zhejiang 314000, China.
| | - W Bian
- Department of Radiology, Jiaxing Maternity and Child Health Care Hospital, Jiaxing, Zhejiang 314000, China
| | - J Chen
- Department of Radiology, Jiaxing Maternity and Child Health Care Hospital, Jiaxing, Zhejiang 314000, China
| | - H Liu
- Department of Radiology, Jiaxing Maternity and Child Health Care Hospital, Jiaxing, Zhejiang 314000, China
| | - Z Wang
- Department of Radiology, Jiaxing Maternity and Child Health Care Hospital, Jiaxing, Zhejiang 314000, China
| |
Collapse
|
196
|
Ferro A, Bottosso M, Dieci MV, Scagliori E, Miglietta F, Aldegheri V, Bonanno L, Caumo F, Guarneri V, Griguolo G, Pasello G. Clinical applications of radiomics and deep learning in breast and lung cancer: A narrative literature review on current evidence and future perspectives. Crit Rev Oncol Hematol 2024; 203:104479. [PMID: 39151838 DOI: 10.1016/j.critrevonc.2024.104479] [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: 01/10/2024] [Revised: 07/22/2024] [Accepted: 08/10/2024] [Indexed: 08/19/2024] Open
Abstract
Radiomics, analysing quantitative features from medical imaging, has rapidly become an emerging field in translational oncology. Radiomics has been investigated in several neoplastic malignancies as it might allow for a non-invasive tumour characterization and for the identification of predictive and prognostic biomarkers. Over the last few years, evidence has been accumulating regarding potential clinical applications of machine learning in many crucial moments of cancer patients' history. However, the incorporation of radiomics in clinical decision-making process is still limited by low data reproducibility and study variability. Moreover, the need for prospective validations and standardizations is emerging. In this narrative review, we summarize current evidence regarding radiomic applications in high-incidence cancers (breast and lung) for screening, diagnosis, staging, treatment choice, response, and clinical outcome evaluation. We also discuss pro and cons of the radiomic approach, suggesting possible solutions to critical issues which might invalidate radiomics studies and propose future perspectives.
Collapse
Affiliation(s)
- Alessandra Ferro
- Division of Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, via Gattamelata 64, Padua 35128, Italy
| | - Michele Bottosso
- Division of Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, via Gattamelata 64, Padua 35128, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padova, via Giustiniani 2, Padova 35128, Italy
| | - Maria Vittoria Dieci
- Division of Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, via Gattamelata 64, Padua 35128, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padova, via Giustiniani 2, Padova 35128, Italy.
| | - Elena Scagliori
- Radiology Unit, Veneto Institute of Oncology IOV - IRCCS, via Gattamelata 64, Padua 35128, Italy
| | - Federica Miglietta
- Division of Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, via Gattamelata 64, Padua 35128, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padova, via Giustiniani 2, Padova 35128, Italy
| | - Vittoria Aldegheri
- Radiology Unit, Veneto Institute of Oncology IOV - IRCCS, via Gattamelata 64, Padua 35128, Italy
| | - Laura Bonanno
- Division of Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, via Gattamelata 64, Padua 35128, Italy
| | - Francesca Caumo
- Unit of Breast Radiology, Veneto Institute of Oncology IOV - IRCCS, via Gattamelata 64, Padua 35128, Italy
| | - Valentina Guarneri
- Division of Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, via Gattamelata 64, Padua 35128, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padova, via Giustiniani 2, Padova 35128, Italy
| | - Gaia Griguolo
- Division of Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, via Gattamelata 64, Padua 35128, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padova, via Giustiniani 2, Padova 35128, Italy
| | - Giulia Pasello
- Division of Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, via Gattamelata 64, Padua 35128, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padova, via Giustiniani 2, Padova 35128, Italy
| |
Collapse
|
197
|
Mayerhoefer ME, Shepherd TM, Weber M, Leithner D, Woo S, Pan JW, Pardoe HR. Sexual Dimorphism of Radiomic Features in the Brain: An Exploratory Study Using 700 μm MP2RAGE MRI at 7 T. Invest Radiol 2024; 59:782-786. [PMID: 38896439 DOI: 10.1097/rli.0000000000001088] [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: 06/21/2024]
Abstract
OBJECTIVES The aim of this study was to determine whether MRI radiomic features of key cerebral structures differ between women and men, and whether detection of such differences depends on the image resolution. MATERIALS AND METHODS Ultrahigh resolution (UHR) 3D MP2RAGE (magnetization-prepared 2 rapid acquisition gradient echo) T1-weighted MR images (voxel size, 0.7 × 0.7 × 0.7 mm 3 ) of the brain of 30 subjects (18 women and 12 men; mean age, 39.0 ± 14.8 years) without abnormal findings on MRI were retrospectively included. MRI was performed on a whole-body 7 T MR system. A convolutional neural network was used to segment the following structures: frontal cortex, frontal white matter, thalamus, putamen, globus pallidus, caudate nucleus, and corpus callosum. Eighty-seven radiomic features were extracted respectively: gray-level histogram (n = 18), co-occurrence matrix (n = 24), run-length matrix (n = 16), size-zone matrix (n = 16), and dependence matrix (n = 13). Feature extraction was performed at UHR and, additionally, also after resampling to 1.4 × 1.4 × 1.4 mm 3 voxel size (standard clinical resolution). Principal components (PCs) of radiomic features were calculated, and independent samples t tests with Cohen d as effect size measure were used to assess differences in PCs between women and men for the different cerebral structures. RESULTS At UHR, at least a single PC differed significantly between women and men in 6/7 cerebral structures: frontal cortex ( d = -0.79, P = 0.042 and d = -1.01, P = 0.010), frontal white matter ( d = -0.81, P = 0.039), thalamus ( d = 1.43, P < 0.001), globus pallidus ( d = 0.92, P = 0.020), caudate nucleus ( d = -0.83, P = 0.039), and corpus callosum ( d = -0.97, P = 0.039). At standard clinical resolution, only a single PC extracted from the corpus callosum differed between sexes ( d = 1.05, P = 0.009). CONCLUSIONS Nonnegligible differences in radiomic features of several key structures of the brain exist between women and men, and need to be accounted for. Very high spatial resolution may be required to uncover and further investigate the sexual dimorphism of brain structures on MRI.
Collapse
Affiliation(s)
- Marius E Mayerhoefer
- From the Department of Radiology, NYU Grossman School of Medicine, New York, NY (M.E.M., T.M.S., D.L., S.W.); Division of General and Pediatric Radiology, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria (M.E.M., M.W.); Florey Institute of Neuroscience and Mental Health, Melbourne, Australia (H.R.P.); Comprehensive Epilepsy Center, Department of Neurology, NYU Grossman School of Medicine, New York, NY (H.R.P.); and Department of Radiology, University of Missouri Columbia, Columbia, MO (J.W.P.)
| | | | | | | | | | | | | |
Collapse
|
198
|
Beddok A, Orlhac F, Rozenblum L, Calugaru V, Créhange G, Dercle L, Nioche C, Thariat J, Marin T, El Fakhri G, Buvat I. Radiomics-driven personalized radiotherapy for primary and recurrent tumors: A general review with a focus on reirradiation. Cancer Radiother 2024; 28:597-602. [PMID: 39406602 DOI: 10.1016/j.canrad.2024.09.002] [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: 08/30/2024] [Accepted: 09/02/2024] [Indexed: 11/03/2024]
Abstract
PURPOSE This review systematically investigates the role of radiomics in radiotherapy, with a particular emphasis on the use of quantitative imaging biomarkers for predicting clinical outcomes, assessing toxicity, and optimizing treatment planning. While the review encompasses various applications of radiomics in radiotherapy, it particularly highlights its potential for guiding reirradiation of recurrent cancers. METHODS A systematic review was conducted based on a Medline search with the search engine PubMed using the keywords "radiomics or radiomic" and "radiotherapy or reirradiation". Out of 189 abstracts reviewed, 147 original articles were included in the analysis. These studies were categorized by tumor localization, imaging modality, study objectives, and performance metrics, with a particular emphasis on the inclusion of external validation and adherence to a standardized radiomics pipeline. RESULTS The review identified 14 tumor localizations, with the majority of studies focusing on lung (33 studies), head and neck (27 studies), and brain (15 studies) cancers. CT was the most frequently employed imaging modality (77 studies) for radiomics, followed by MRI (46 studies) and PET (13 studies). The overall AUC across all studies, primarily focused on predicting the risk of recurrence (94 studies) or toxicity (41 studies), was 0.80 (SD=0.08). However, only 24 studies (16.3%) included external validation, with a slightly lower AUC compared to those without it. For studies using CT versus MRI or PET, both had a median AUC of 0.79, with IQRs of 0.73-0.86 for CT and 0.76-0.855 for MRI/PET, showing no significant differences in performance. Five studies involving reirradiation reported a median AUC of 0.81 (IQR: 0.73-0.825). CONCLUSION Radiomics demonstrates considerable potential in personalizing radiotherapy by improving treatment precision through better outcome prediction and treatment planning. However, its clinical adoption is hindered by the lack of external validation and variability in study designs. Future research should focus on implementing rigorous validation methods and standardizing imaging protocols to enhance the reliability and generalizability of radiomics in clinical radiotherapy, with particular attention to its application in reirradiation.
Collapse
Affiliation(s)
- Arnaud Beddok
- Department of Radiation Oncology, institut Godinot, Reims, France; Université de Reims Champagne-Ardenne, Crestic, Reims, France; Yale PET Center, Department of Radiology & Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut, USA.
| | - Fanny Orlhac
- Institut Curie, université PSL, université Paris Saclay, Inserm Lito U1288, Orsay, France
| | - Laura Rozenblum
- Yale PET Center, Department of Radiology & Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut, USA; Department of Nuclear Medicine, hôpitaux universitaires la Pitié Salpêtrière-Charles-Foix, AP-HP, Sorbonne université, 75013 Paris, France
| | - Valentin Calugaru
- Institut Curie, université PSL, université Paris Saclay, Inserm Lito U1288, Orsay, France; Department of Radiation Oncology, institut Curie, université PSL, Paris, France
| | - Gilles Créhange
- Institut Curie, université PSL, université Paris Saclay, Inserm Lito U1288, Orsay, France; Department of Radiation Oncology, institut Curie, université PSL, Paris, France
| | - Laurent Dercle
- Department of Radiology, New York-Presbyterian Hospital, Columbia University, Vagelos College of Physicians and Surgeons, New York, USA
| | - Christophe Nioche
- Institut Curie, université PSL, université Paris Saclay, Inserm Lito U1288, Orsay, France
| | - Juliette Thariat
- Department of Radiation Oncology, centre François-Baclesse, Caen, France
| | - Thibault Marin
- Yale PET Center, Department of Radiology & Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Georges El Fakhri
- Yale PET Center, Department of Radiology & Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Irène Buvat
- Institut Curie, université PSL, université Paris Saclay, Inserm Lito U1288, Orsay, France
| |
Collapse
|
199
|
Williams MC, Weir-McCall JR, Baldassarre LA, De Cecco CN, Choi AD, Dey D, Dweck MR, Isgum I, Kolossvary M, Leipsic J, Lin A, Lu MT, Motwani M, Nieman K, Shaw L, van Assen M, Nicol E. Artificial Intelligence and Machine Learning for Cardiovascular Computed Tomography (CCT): A White Paper of the Society of Cardiovascular Computed Tomography (SCCT). J Cardiovasc Comput Tomogr 2024; 18:519-532. [PMID: 39214777 DOI: 10.1016/j.jcct.2024.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 08/03/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024]
Affiliation(s)
| | | | - Lauren A Baldassarre
- Section of Cardiovascular Medicine and Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
| | | | - Andrew D Choi
- The George Washington University School of Medicine, Washington, USA
| | - Damini Dey
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, USA
| | - Marc R Dweck
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Ivana Isgum
- Amsterdam University Medical Center, University of Amsterdam, Netherlands
| | - Márton Kolossvary
- Gottsegen National Cardiovascular Center, Budapest, Hungary, and Physiological Controls Research Center, University Research and Innovation Center, Óbuda University, Budapest, Hungary
| | | | - Andrew Lin
- Victorian Heart Institute and Monash Health Heart, Victorian Heart Hospital, Monash University, Australia
| | - Michael T Lu
- Massachusetts General Hospital Cardiovascular Imaging Research Center/Harvard Medical School, USA
| | | | | | - Leslee Shaw
- Icahn School of Medicine at Mount Sinai, New York, USA
| | | | - Edward Nicol
- Royal Brompton Hospital, Guys and St Thomas' NHS Foundation Trust, London, UK; School of Biomedical Engineering and Imaging Sciences, King's College London, UK
| |
Collapse
|
200
|
Sun C, Fan E, Huang L, Zhang Z. Performance of radiomics in preoperative determination of malignant potential and Ki-67 expression levels in gastrointestinal stromal tumors: a systematic review and meta-analysis. Acta Radiol 2024; 65:1307-1318. [PMID: 39411915 DOI: 10.1177/02841851241285958] [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] [Indexed: 11/13/2024]
Abstract
Empirical evidence for radiomics predicting the malignant potential and Ki-67 expression in gastrointestinal stromal tumors (GISTs) is lacking. The aim of this review article was to explore the preoperative discriminative performance of radiomics in assessing the malignant potential, mitotic index, and Ki-67 expression levels of GISTs. We systematically searched PubMed, EMBASE, Web of Science, and the Cochrane Library. The search was conducted up to 30 September 2023. Quality assessment was performed using the Radiomics Quality Score (RQS). A total of 35 original studies were included in the analysis. Among them, 26 studies focused on determining malignant potential, three studies on mitotic index discrimination, and six studies on Ki-67 discrimination. In the validation set, the sensitivity, specificity, and area under the receiver operating characteristic curve (AUC) of radiomics in the determination of high malignant potential were 0.74 (95% CI=0.69-0.78), 0.90 (95% CI=0.83-0.94), and 0.81 (95% CI=0.14-0.99), respectively. For moderately to highly malignant potential, the sensitivity, specificity, and AUC were 0.86 (95% CI=0.83-0.88), 0.73 (95% CI=0.67-0.78), and 0.88 (95% CI=0.27-0.99), respectively. Regarding the determination of high mitotic index, the sensitivity, specificity, and AUC of radiomics were 0.86 (95% CI=0.83-0.88), 0.73 (95% CI=0.67-0.78), and 0.88 (95% CI=0.27-0.99), respectively. When determining high Ki-67 expression, the combined sensitivity, specificity, and AUC were 0.74 (95% CI=0.65-0.81), 0.81 (95% CI=0.74-0.86), and 0.84 (95% CI=0.61-0.95), respectively. Radiomics demonstrates promising discriminative performance in the preoperative assessment of malignant potential, mitotic index, and Ki-67 expression levels in GISTs.
Collapse
Affiliation(s)
- Chengyu Sun
- Department of Colorectal Surgery, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, Jiangsu, PR China
| | - Enguo Fan
- State Key Laboratory of Medical Molecular Biology, Department of Microbiology and Parasitology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, PR China
| | - Luqiao Huang
- Department of Colorectal Surgery, Xuzhou Central Hospital, Xuzhou, Jiangsu, PR China
| | - Zhengguo Zhang
- Department of Colorectal Surgery, Xuzhou Central Hospital, Xuzhou, Jiangsu, PR China
| |
Collapse
|