1
|
Huang S, Liu D, Deng K, Shu C, Wu Y, Zhou Z. A computed tomography angiography-based radiomics model for prognostic prediction of endovascular abdominal aortic repair. Int J Cardiol 2025; 429:133138. [PMID: 40090490 DOI: 10.1016/j.ijcard.2025.133138] [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: 01/21/2025] [Revised: 02/25/2025] [Accepted: 03/06/2025] [Indexed: 03/18/2025]
Abstract
OBJECTIVE This study aims to develop a radiomics machine learning (ML) model that uses preoperative computed tomography angiography (CTA) data to predict the prognosis of endovascular aneurysm repair (EVAR) for abdominal aortic aneurysm (AAA) patients. METHODS In this retrospective study, 164 AAA patients underwent EVAR and were categorized into shrinkage (good prognosis) or stable (poor prognosis) groups based on post-EVAR sac regression. From preoperative AAA and perivascular adipose tissue (PVAT) image, radiomics features (RFs) were extracted for model creation. Patients were split into 80 % training and 20 % test sets. A support vector machine model was constructed for prediction. Accuracy is evaluated via the area under the receiver operating characteristic curve (AUC). RESULTS Demographics and comorbidities showed no significant differences between shrinkage and stable groups. The model containing 5 AAA RFs (which are original_firstorder_InterquartileRange, log-sigma-3-0-mm-3D_glrlm_GrayLevelNonUniformityNormalized, log-sigma-3-0-mm-3D_glrlm_RunPercentage, log-sigma-4-0-mm-3D_glrlm_ShortRunLowGrayLevelEmphasis, wavelet-LLH_glcm_SumEntropy) had AUCs of 0.86 (training) and 0.77 (test). The model containing 7 PVAT RFs (which are log-sigma-3-0-mm-3D_firstorder_InterquartileRange, log-sigma-3-0-mm-3D_glcm_Correlation, wavelet-LHL_firstorder_Energy, wavelet-LHL_firstorder_TotalEnergy, wavelet-LHH_firstorder_Mean, wavelet-LHH_glcm_Idmn, wavelet-LHH_glszm_GrayLevelNonUniformityNormalized) had AUCs of 0.76 (training) and 0.78 (test). Combining AAA and PVAT RFs yielded the highest accuracy: AUCs of 0.93 (training) and 0.87 (test). CONCLUSIONS Radiomics-based CTA model predicts aneurysm sac regression post-EVAR in AAA patients. PVAT RFs from preoperative CTA images were closely related to AAA prognosis after EVAR, enhancing accuracy when combined with AAA RFs. This preliminary study explores a predictive model designed to assist clinicians in optimizing therapeutic strategies during clinical decision-making processes.
Collapse
Affiliation(s)
- Shanya Huang
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, 139 Middle Renmin Road, Changsha, 410011, Hunan, China; Department of Ultrasound, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Dingxiao Liu
- Department of Vascular Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Kai Deng
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Chang Shu
- Department of Vascular Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China; Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Yan Wu
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, 139 Middle Renmin Road, Changsha, 410011, Hunan, China.
| | - Zhiguang Zhou
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, 139 Middle Renmin Road, Changsha, 410011, Hunan, China.
| |
Collapse
|
2
|
Benhabib H, Brandenberger D, Lajkosz K, Demicco EG, Tsoi KM, Wunder JS, Ferguson PC, Griffin AM, Naraghi A, Haider MA, White LM. MRI Radiomics Analysis in the Diagnostic Differentiation of Malignant Soft Tissue Myxoid Sarcomas From Benign Soft Tissue Musculoskeletal Myxomas. J Magn Reson Imaging 2025; 61:2630-2641. [PMID: 39843987 PMCID: PMC12063761 DOI: 10.1002/jmri.29691] [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: 10/01/2024] [Revised: 12/11/2024] [Accepted: 12/12/2024] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND Differentiation of benign myxomas and malignant myxoid sarcomas can be difficult with an overlapping spectrum of morphologic MR findings. PURPOSE To assess the diagnostic utility of MRI radiomics in the differentiation of musculoskeletal myxomas and myxoid sarcomas. STUDY TYPE Retrospective. POPULATION A total of 523 patients were included; histologically proven myxomas (N = 201) and myxoid sarcomas (N = 322), randomly divided (70:30) into training:test subsets. SEQUENCE/FIELD STRENGTH T1-weighted (T1W), T2-weighted fat-suppressed (fluid-sensitive), and T1-weighted post-contrast (T1W + C) sequences at 1.0 T, 1.5 T, or 3.0 T. ASSESSMENT Seven semantic (qualitative) tumor features were assessed in each case. Manual 3D tumor segmentations performed with radiomics features extracted from T1W, fluid-sensitive, and T1W + C acquisitions. Models were constructed based on radiomic features from individual sequences and from their combination, both with and without the addition of qualitative tumor features. STATISTICAL TESTS Intraclass correlation evaluated in 60 cases segmented by three readers. Features with intraclass correlation <0.7 excluded from further analysis. Boruta feature selection and Random Forest modeling performed using the training-dataset, with resultant models used to assess class discrimination (myxoma vs. myxoid sarcoma) in the test dataset. Radiomics score defined as probability class = myxoma. Logistic regression modeling employed to estimate performance of the radiomics score. Area under the receiver operating characteristic curve (AUC) was used to assess diagnostic performance, and DeLong's test to assess performance between constructed models. A P-value <0.05 was considered significant. RESULTS Four qualitative semantic features showed significant predictive power in class discrimination. Radiomic models demonstrated excellent differentiation of myxomas from myxoid sarcomas: AUC of 0.9271 (T1W), 0.9049 (fluid-sensitive), and 0.9179 (T1W + C). Incorporation of multiparametric data or semantic features did not significantly improve model performance (P ≥ 0.08) compared to radiomic models derived from any individual MRI sequence alone. DATA CONCLUSION MRI radiomics appears to be accurate in the differentiation of myxomas from myxoid sarcomas. Classification performance did not improve when incorporating qualitative features or multiparametric imaging data. PLAIN LANGUAGE SUMMARY Accurately distinguishing between benign soft tissue myxomas and malignant myxoid sarcomas is essential for guiding appropriate management but remains challenging with conventional MRI interpretation. This study utilized radiomics, a method that extracts quantitative mathematically derived features from images, to develop predictive models based on routine MRI examination. Analyzing over 500 cases, MRI radiomics demonstrated excellent diagnostic accuracy in differentiating between benign myxomas and malignant myxoid sarcomas, highlighting the potential of the technique, as a powerful non-invasive tool that could complement current diagnostic approaches, and enhance clinical decision-making in patients with soft tissue myxoid tumors of the musculoskeletal system. LEVEL OF EVIDENCE 3 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Hadas Benhabib
- Department of Medical ImagingUniversity of TorontoTorontoOntarioCanada
- Joint Department of Medical ImagingUniversity Health Network, Sinai Health System, Women's College Hospital, Mount Sinai HospitalTorontoOntarioCanada
| | - Daniel Brandenberger
- Department of Medical ImagingUniversity of TorontoTorontoOntarioCanada
- Joint Department of Medical ImagingUniversity Health Network, Sinai Health System, Women's College Hospital, Mount Sinai HospitalTorontoOntarioCanada
- Institut für Radiologie und NuklearmedizinKantonsspital BasellandLiestalSwitzerland
| | - Katherine Lajkosz
- Department of BiostatisticsUniversity Health NetworkTorontoOntarioCanada
| | - Elizabeth G. Demicco
- Department of Pathology and Laboratory MedicineMount Sinai HospitalTorontoOntarioCanada
- Department of Laboratory Medicine and PathobiologyUniversity of TorontoTorontoOntarioCanada
| | - Kim M. Tsoi
- Department of Pathology and Laboratory MedicineMount Sinai HospitalTorontoOntarioCanada
- Department of Laboratory Medicine and PathobiologyUniversity of TorontoTorontoOntarioCanada
- Department of SurgeryUniversity of TorontoTorontoOntarioCanada
| | - Jay S. Wunder
- Department of SurgeryUniversity of TorontoTorontoOntarioCanada
- University Musculoskeletal Oncology Unit, Division of Orthopedic SurgeryMount Sinai HospitalTorontoOntarioCanada
| | - Peter C. Ferguson
- Department of SurgeryUniversity of TorontoTorontoOntarioCanada
- University Musculoskeletal Oncology Unit, Division of Orthopedic SurgeryMount Sinai HospitalTorontoOntarioCanada
| | - Anthony M. Griffin
- University Musculoskeletal Oncology Unit, Division of Orthopedic SurgeryMount Sinai HospitalTorontoOntarioCanada
| | - Ali Naraghi
- Department of Medical ImagingUniversity of TorontoTorontoOntarioCanada
- Joint Department of Medical ImagingUniversity Health Network, Sinai Health System, Women's College Hospital, Mount Sinai HospitalTorontoOntarioCanada
| | - Masoom A. Haider
- Department of Medical ImagingUniversity of TorontoTorontoOntarioCanada
- Joint Department of Medical ImagingUniversity Health Network, Sinai Health System, Women's College Hospital, Mount Sinai HospitalTorontoOntarioCanada
| | - Lawrence M. White
- Department of Medical ImagingUniversity of TorontoTorontoOntarioCanada
- Joint Department of Medical ImagingUniversity Health Network, Sinai Health System, Women's College Hospital, Mount Sinai HospitalTorontoOntarioCanada
- Department of SurgeryUniversity of TorontoTorontoOntarioCanada
| |
Collapse
|
3
|
Fang Y, Zhang Q, Yan J, Yu S. Application of radiomics in acute and severe non-neoplastic diseases: A literature review. J Crit Care 2025; 87:155027. [PMID: 39848114 DOI: 10.1016/j.jcrc.2025.155027] [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: 05/14/2024] [Revised: 11/01/2024] [Accepted: 01/10/2025] [Indexed: 01/25/2025]
Abstract
Radiomics involves the integration of computer technology, big data analysis, and clinical medicine. Currently, there have been initial advancements in the fields of acute cerebrovascular disease and cardiovascular disease. The objective of radiomics is to extract quantitative features from medical images for analysis to predict the risk or treatment outcome, help in differential diagnosis, and guide clinical decisions and management. Radiomics applied research has reached a more advanced stage yet encounters several obstacles, including the need for standardization of radiomics features and alignment with treatment requirements for acute and severe illnesses. Future research should aim to seamlessly incorporate radiomics with various disciplines, leverage big data and artificial intelligence advancements, cater to the requirements of acute and critical medicine, and enhance the effectiveness of technological innovation and application in diagnosing and treating acute and critical illnesses.
Collapse
Affiliation(s)
- Yu Fang
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China; Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Qiannan Zhang
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Jingjun Yan
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China; Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Shanshan Yu
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China; Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China.
| |
Collapse
|
4
|
Xia Y, Sun Z, Wang Z, Zhang X, Xu J, Li M, Mao N, Xu C, Li X, Xu H, Yang Z, Ma H, Guo H. Intra- and Peritumoral CT-Based Radiomics for Assessing Pathologic T-Staging in Clear Cell Renal Cell Carcinoma: A Multicenter Study. Ann Surg Oncol 2025; 32:4550-4561. [PMID: 40106107 DOI: 10.1245/s10434-025-17111-4] [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: 11/04/2024] [Accepted: 02/17/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND A radiomics model constructed from the intratumoral region of computed tomography (CT) can predict the pathologic T stage of clear cell renal cell carcinoma (ccRCC). However, the predictive capability of the radiomics model that incorporates both intra- and peritumoral regions of CT for the pathologic T stage in ccRCC patients has not been reported to date. METHODS This study enrolled 250 patients with ccRCC who underwent laparoscopic surgery. Three radiomics models were developed based on the intra- and peritumoral regions. The sensitivity, specificity, accuracy, and receiver operating characteristic (ROC) curves of each model were analyzed. Decision curve analysis (DCA) and calibration curves were used to assess the net benefit and calibration ability of the models. Additionally, the diagnostic performance of the different models were compared with that of radiologists. RESULTS The radiomics model based on the intra- and peritumoral regions at 5 mm exhibited the strongest performance, with area under curve values of 0.91 (95 % confidence interval [CI], 0.8551-0.9650), 0.85 (95 % CI, 0.7490-0.9517), and 0.873 (95 % CI, 0.7612-0.9839) in distinguishing high and low T stages of ccRCC across the training, validation, and test sets, respectively. The model's accuracy in the training, validation, and test sets was 0.798, 0.732, and 0.769, with corresponding sensitivity values of 0.921, 0.857, and 0.882, and specificity values of 0.747, 0.690, and 0.729. The calibration curve demonstrated a high level of agreement between the predicted and actual outcomes, whereas the DCA showed that the model provided a meaningful net benefit. CONCLUSIONS The radiomics model based on the intra- and peritumoral regions of CT has certain value in distinguishing between high and low T stages of ccRCC.
Collapse
Affiliation(s)
- Yuanhao Xia
- School of Medical Imaging, Binzhou Medical University, Yantai, China
- Department of Radiology, Qingdao University Medical College Affiliated Yantai Yuhuangding Hospital, Yantai, China
| | - Zehua Sun
- Department of Radiology, The First Hospital of Jilin University, Changchun, China
| | - Zhongyi Wang
- Department of Radiology, Qingdao University Medical College Affiliated Yantai Yuhuangding Hospital, Yantai, China
| | - Xin Zhang
- Department of Personnel, Lianshui County People's Hospital, Huai'an, China
| | - Jiakang Xu
- Department of Radiology, Qingdao University Medical College Affiliated Yantai Yuhuangding Hospital, Yantai, China
| | - Min Li
- School of Medical Imaging, Binzhou Medical University, Yantai, China
| | - Ning Mao
- Department of Radiology, Qingdao University Medical College Affiliated Yantai Yuhuangding Hospital, Yantai, China
| | - Chang Xu
- School of Medical Imaging, Binzhou Medical University, Yantai, China
| | - Xianglin Li
- School of Medical Imaging, Binzhou Medical University, Yantai, China
| | - Hui Xu
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zhenghan Yang
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Heng Ma
- School of Medical Imaging, Binzhou Medical University, Yantai, China
- Department of Radiology, Qingdao University Medical College Affiliated Yantai Yuhuangding Hospital, Yantai, China
| | - Hao Guo
- School of Medical Imaging, Binzhou Medical University, Yantai, China.
- Department of Radiology, Qingdao University Medical College Affiliated Yantai Yuhuangding Hospital, Yantai, China.
| |
Collapse
|
5
|
Zhu Y, Liu T, Chen J, Wen L, Zhang J, Zheng D. Prediction of therapeutic response to transarterial chemoembolization plus systemic therapy regimen in hepatocellular carcinoma using pretreatment contrast-enhanced MRI based habitat analysis and Crossformer model. Abdom Radiol (NY) 2025; 50:2464-2475. [PMID: 39586897 DOI: 10.1007/s00261-024-04709-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 11/14/2024] [Accepted: 11/15/2024] [Indexed: 11/27/2024]
Abstract
PURPOSE To develop habitat and deep learning (DL) models from multi-phase contrast-enhanced magnetic resonance imaging (CE-MRI) habitat images categorized using the K-means clustering algorithm. Additionally, we aim to assess the predictive value of identified regions for early evaluation of the responsiveness of hepatocellular carcinoma (HCC) patients to treatment with transarterial chemoembolization (TACE) plus molecular targeted therapies (MTT) and anti-PD-(L)1. METHODS A total of 102 patients with HCC from two institutions (A, n = 63 and B, n = 39) who received TACE plus systemic therapy were enrolled from September 2020 to January 2024. Multiple CE-MRI sequences were used to outline 3D volumes of interest (VOI) of the lesion. Subsequently, K-means clustering was applied to categorize intratumoral voxels into three distinct subgroups, based on signal intensity values of images. Using data from institution A, the habitat model was built with the ExtraTrees classifier after extracting radiomics features from intratumoral habitats. Similarly, the Crossformer model and ResNet50 model were trained on multi-channel data in institution A, and a DL model with Transformer-based aggregation was constructed to predict the response. Finally, all models underwent validation at institution B. RESULTS The Crossformer model and the habitat model both showed high area under the receiver operating characteristic curves (AUCs) of 0.869 and 0.877 (training cohort). In validation, AUC was 0.762 for the Crossformer model and 0.721 for the habitat model. CONCLUSION The habitat model and DL model based on CE-MRI possesses the capability to non-invasively predict the efficacy of TACE plus systemic therapy in HCC patients, which is critical for precision treatment and patient outcomes.
Collapse
Affiliation(s)
- Yuemin Zhu
- Department of Radiology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Tao Liu
- Department of Radiology, Chongqing University Cancer Hospital, Chongqing Cancer Institute, and Chongqing Cancer Hospital, Chongqing, China
| | - Jianwei Chen
- Department of Radiology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Liting Wen
- Department of Radiology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Jiuquan Zhang
- Department of Radiology, Chongqing University Cancer Hospital, Chongqing Cancer Institute, and Chongqing Cancer Hospital, Chongqing, China.
| | - Dechun Zheng
- Department of Radiology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China.
| |
Collapse
|
6
|
Wang ZD, Nan HJ, Li SX, Li LH, Liu ZC, Guo HH, Li L, Liu SY, Li H, Bai YL, Dang XW. Development and validation of a radiomics-based prediction model for variceal bleeding in patients with Budd-Chiari syndrome-related gastroesophageal varices. World J Gastroenterol 2025; 31:104563. [DOI: 10.3748/wjg.v31.i19.104563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 03/24/2025] [Accepted: 04/27/2025] [Indexed: 05/21/2025] Open
Abstract
BACKGROUND Budd-Chiari syndrome (BCS) is caused by obstruction of the hepatic veins or suprahepatic inferior vena cava, leading to portal hypertension and the development of gastroesophageal varices (GEVs), which are associated with an increased risk of bleeding. Existing risk models for variceal bleeding in cirrhotic patients have limited applicability to BCS due to differences in pathophysiology. Radiomics, as a noninvasive technique, holds promise as a tool for more accurate prediction of bleeding risk in BCS-related GEVs.
AIM To develop and validate a personalized risk model for predicting variceal bleeding in BCS patients with GEVs.
METHODS We retrospectively analyzed clinical data from 444 BCS patients with GEVs in two centers. Radiomic features were extracted from portal venous phase computed tomography (CT) scans. A training cohort of 334 patients was used to develop the model, with 110 patients serving as an external validation cohort. LASSO Cox regression was used to select radiomic features for constructing a radiomics score (Radscore). Univariate and multivariate Cox regression identified independent clinical predictors. A combined radiomics + clinical (R + C) model was developed using stepwise regression. Model performance was assessed using the area under the receiver operating characteristic curve (AUC), calibration plots, and decision curve analysis (DCA), with external validation to evaluate generalizability.
RESULTS The Radscore comprised four hepatic and six splenic CT features, which predicted the risk of variceal bleeding. Multivariate analysis identified invasive treatment to relieve hepatic venous outflow obstruction, anticoagulant therapy, and hemoglobin levels as independent clinical predictors. The R + C model achieved C-indices of 0.906 (training) and 0.859 (validation), outperforming the radiomics and clinical models alone (AUC: training 0.936 vs 0.845 vs 0.823; validation 0.876 vs 0.712 vs 0.713). DCA showed higher clinical net benefit across the thresholds. The model stratified patients into low-, medium- and high-risk groups with significant differences in bleeding rates (P < 0.001). An online tool is available at https://bcsvh.shinyapps.io/BCS_Variceal_Bleeding_Risk_Tool/.
CONCLUSION We developed and validated a novel radiomics-based model that noninvasively and conveniently predicted risk of variceal bleeding in BCS patients with GEVs, aiding early identification and management of high-risk patients.
Collapse
Affiliation(s)
- Ze-Dong Wang
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
- Key Laboratory of Precision Diagnosis and Treatment in General Surgical (Hepatobiliary and Pancreatic) Diseases of Health Commission of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
- Henan Province Engineering Research Center of Minimally Invasive Diagnosis and Treatment of Hepatobiliary and Pancreatic Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
- Budd-Chiari Syndrome Diagnosis and Treatment Center of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Hui-Jie Nan
- Department of Hematology, Zhengzhou University People's Hospital and Henan Provincial People's Hospital, Zhengzhou 450003, Henan Province, China
| | - Su-Xin Li
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
- Key Laboratory of Precision Diagnosis and Treatment in General Surgical (Hepatobiliary and Pancreatic) Diseases of Health Commission of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
- Henan Province Engineering Research Center of Minimally Invasive Diagnosis and Treatment of Hepatobiliary and Pancreatic Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
- Budd-Chiari Syndrome Diagnosis and Treatment Center of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Lu-Hao Li
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
- Key Laboratory of Precision Diagnosis and Treatment in General Surgical (Hepatobiliary and Pancreatic) Diseases of Health Commission of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
- Henan Province Engineering Research Center of Minimally Invasive Diagnosis and Treatment of Hepatobiliary and Pancreatic Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
- Budd-Chiari Syndrome Diagnosis and Treatment Center of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Zhao-Chen Liu
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
- Key Laboratory of Precision Diagnosis and Treatment in General Surgical (Hepatobiliary and Pancreatic) Diseases of Health Commission of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
- Henan Province Engineering Research Center of Minimally Invasive Diagnosis and Treatment of Hepatobiliary and Pancreatic Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
- Budd-Chiari Syndrome Diagnosis and Treatment Center of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Hua-Hu Guo
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
- Key Laboratory of Precision Diagnosis and Treatment in General Surgical (Hepatobiliary and Pancreatic) Diseases of Health Commission of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
- Henan Province Engineering Research Center of Minimally Invasive Diagnosis and Treatment of Hepatobiliary and Pancreatic Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
- Budd-Chiari Syndrome Diagnosis and Treatment Center of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Lin Li
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
- Key Laboratory of Precision Diagnosis and Treatment in General Surgical (Hepatobiliary and Pancreatic) Diseases of Health Commission of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
- Henan Province Engineering Research Center of Minimally Invasive Diagnosis and Treatment of Hepatobiliary and Pancreatic Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
- Budd-Chiari Syndrome Diagnosis and Treatment Center of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Sheng-Yan Liu
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
- Key Laboratory of Precision Diagnosis and Treatment in General Surgical (Hepatobiliary and Pancreatic) Diseases of Health Commission of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
- Henan Province Engineering Research Center of Minimally Invasive Diagnosis and Treatment of Hepatobiliary and Pancreatic Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
- Budd-Chiari Syndrome Diagnosis and Treatment Center of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Hai Li
- Department of Hepatopancreatobiliary Surgery, Zhengzhou University People's Hospital and Henan Provincial People's Hospital, Zhengzhou 450003, Henan Province, China
| | - Yan-Liang Bai
- Department of Hematology, Zhengzhou University People's Hospital and Henan Provincial People's Hospital, Zhengzhou 450003, Henan Province, China
| | - Xiao-Wei Dang
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
- Key Laboratory of Precision Diagnosis and Treatment in General Surgical (Hepatobiliary and Pancreatic) Diseases of Health Commission of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
- Henan Province Engineering Research Center of Minimally Invasive Diagnosis and Treatment of Hepatobiliary and Pancreatic Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
- Budd-Chiari Syndrome Diagnosis and Treatment Center of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| |
Collapse
|
7
|
Xiao S, Zeng S, Kou Y. MRI radiomics in diagnosing high and low grade meningiomas through systematic review and meta analysis. Sci Rep 2025; 15:17521. [PMID: 40394344 DOI: 10.1038/s41598-025-88315-7] [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: 02/29/2024] [Accepted: 01/28/2025] [Indexed: 05/22/2025] Open
Abstract
To evaluate the diagnostic value of magnetic resonance imaging (MRI) radiomics in distinguishing high-grade meningiomas (HGM) from low-grade meningiomas (LGM). A systematic search was conducted in PubMed, EMbase, Web of Science, and The Cochrane Library databases up to December 31, 2023. Two researchers independently screened studies, extracted data, and assessed risk of bias and quality of included studies as well. Meta-analysis was performed using Stata 14 software to calculate pooled sensitivity (SEN), specificity (SPE), positive likelihood ratio (PLR), negative likelihood ratio (NLR), diagnostic odds ratio (DOR), and area under the curve (AUC). A total of 21 studies with 2253 patients were included (607 HGM, 1646 LGM). Meta-analysis showed an overall SEN of 0.82 (95% CI 0.74-0.88) and SPE of 0.85 (95% CI 0.81-0.89). The PLR and NLR were 5.64 (95% CI 4.17-7.64) and 0.21 (95% CI 0.14-0.31), respectively, with a pooled DOR of 26.66 (95% CI 14.42-49.27) and an AUC of 0.91 (95% CI 0.88-0.93), indicating high diagnostic accuracy. Although additional research is required to validate suitable techniques, MRI radiomics shows strong potential as an accurate tool for meningioma grading. Standardizing radiomics application could enhance diagnostic precision and clinical decision-making for meningioma grading in the future.Trial Registration: CRD42024500086.
Collapse
Affiliation(s)
- Simin Xiao
- Radiology Department, The First Affiliated Hospital of Traditional Chinese Medicine of Chengdu Medical College, XinDu Hospital of Traditional Chinese Medicine, Chengdu, China
| | - Siyuan Zeng
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yangbin Kou
- Radiology Department, The First Affiliated Hospital of Traditional Chinese Medicine of Chengdu Medical College, XinDu Hospital of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
8
|
Chen B, Song Y, Wang H, Tang L, Xie X, Mao A, Chen Q, Song B. MRI-based model to predict preoperative extrathyroidal extension in papillary thyroid carcinoma. Eur Radiol 2025:10.1007/s00330-025-11684-0. [PMID: 40382730 DOI: 10.1007/s00330-025-11684-0] [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: 02/08/2025] [Revised: 03/26/2025] [Accepted: 04/19/2025] [Indexed: 05/20/2025]
Abstract
OBJECTIVE This study aimed to develop and validate a predictive model for preoperative extrathyroidal extension (ETE) in papillary thyroid carcinoma (PTC) using MRI features. METHODS We retrospectively analyzed 140 confirmed PTC cases, divided into training (n = 84) and validation (n = 56) groups. MRI features such as T2-weighted imaging, multiphase contrast-enhanced MRI, and diffusion-weighted imaging were evaluated along with clinical data. Univariate and multivariate logistic regression identified independent predictors of ETE and developed a predictive nomogram. We evaluated the nomogram's discrimination, calibration, and clinical utility, and performed subgroup analyses to explore the relationships between risk factors and baseline data. Predictive performance was assessed using ROC curves and DeLong tests. RESULTS Age, protrusion value, and apparent diffusion coefficient_Brightest_rate (ADC_Best_rate) were independent predictors of ETE. The nomogram effectively differentiated ETE from no-ETE, showing strong discrimination, clinical utility, and calibration in both the training (AUC = 0.826, Hosmer-Lemeshow p = 0.882) and validation cohorts (AUC = 0.805, Hosmer-Lemeshow p = 0.585). The model performed consistently across different MRI systems (1.5 T and 3.0 T) and gender subgroups. Notably, ADC_Best_rate (AUC = 0.742) outperformed ADC_mean_rate and ADC_minimum_rate. A significant interaction between ADC_Best_rate and gender (p = 0.02) showed that ADC_Best_rate predicted ETE in PTC more accurately in males (AUC = 0.897) compared to females (AUC = 0.644). CONCLUSION Our nomogram model, incorporating age, protrusion value, and ADC_Best_rate, effectively predicted preoperative ETE in PTC patients, aiding surgeons in optimizing therapeutic decision-making. ADC_Best_rate may be a promising potential indicator in MRI functional imaging. KEY POINTS Question This study addresses the challenge of accurately predicting extrathyroidal extension (ETE) in papillary thyroid carcinoma (PTC) to improve surgical decision-making. Findings A predictive nomogram incorporating age, protrusion value, and ADC_Best_rate effectively differentiates ETE from no-ETE, showing strong performance in both training and validation cohorts. Clinical relevance This nomogram aids surgeons in identifying patients at risk for ETE, enhancing therapeutic decision-making and potentially improving patient outcomes in PTC management.
Collapse
Affiliation(s)
- Biaoling Chen
- Department of Radiology, Minhang Hospital, Fudan University, Shanghai, China
| | - Yining Song
- Shanghai Medical College, Fudan University, Shanghai, China
| | - Hao Wang
- Department of Radiology, Minhang Hospital, Fudan University, Shanghai, China
| | - Lang Tang
- Department of Ultrasound, Minhang Hospital, Fudan University, Shanghai, China
| | - Xiaoli Xie
- Department of Pathology, Minhang Hospital, Fudan University, Shanghai, China
| | - Anwei Mao
- Department of General Surgery, Minhang Hospital, Fudan University, Shanghai, China
| | - Qiaohui Chen
- Department of Radiology, Minhang Hospital, Fudan University, Shanghai, China.
| | - Bin Song
- Department of Radiology, Minhang Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
9
|
Yang H, Zhang Y, Li F, Liu W, Zeng H, Yuan H, Ye Z, Huang Z, Yuan Y, Xiang Y, Wu K, Liu H. CT-based AI framework leveraging multi-scale features for predicting pathological grade and Ki67 index in clear cell renal cell carcinoma: a multicenter study. Insights Imaging 2025; 16:102. [PMID: 40369234 PMCID: PMC12078187 DOI: 10.1186/s13244-025-01980-0] [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: 07/22/2024] [Accepted: 03/30/2025] [Indexed: 05/16/2025] Open
Abstract
PURPOSE To explore whether a CT-based AI framework, leveraging multi-scale features, can offer a non-invasive approach to accurately predict pathological grade and Ki67 index in clear cell renal cell carcinoma (ccRCC). METHODS In this multicenter retrospective study, a total of 1073 pathologically confirmed ccRCC patients from seven cohorts were split into internal cohorts (training and validation sets) and an external test set. The AI framework comprised an image processor, a 3D-kidney and tumor segmentation model by 3D-UNet, a multi-scale features extractor built upon unsupervised learning, and a multi-task classifier utilizing XGBoost. A quantitative model interpretation technique, known as SHapley Additive exPlanations (SHAP), was employed to explore the contribution of multi-scale features. RESULTS The 3D-UNet model showed excellent performance in segmenting both the kidney and tumor regions, with Dice coefficients exceeding 0.92. The proposed multi-scale features model exhibited strong predictive capability for pathological grading and Ki67 index, with AUROC values of 0.84 and 0.87, respectively, in the internal validation set, and 0.82 and 0.82, respectively, in the external test set. The SHAP results demonstrated that features from radiomics, the 3D Auto-Encoder, and dimensionality reduction all made significant contributions to both prediction tasks. CONCLUSIONS The proposed AI framework, leveraging multi-scale features, accurately predicts the pathological grade and Ki67 index of ccRCC. CRITICAL RELEVANCE STATEMENT The CT-based AI framework leveraging multi-scale features offers a promising avenue for accurately predicting the pathological grade and Ki67 index of ccRCC preoperatively, indicating a direction for non-invasive assessment. KEY POINTS Non-invasively determining pathological grade and Ki67 index in ccRCC could guide treatment decisions. The AI framework integrates segmentation, classification, and model interpretation, enabling fully automated analysis. The AI framework enables non-invasive preoperative detection of high-risk tumors, assisting clinical decision-making.
Collapse
Affiliation(s)
- Huancheng Yang
- Department of Radiology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Department of Radiology, The Third Affiliated Hospital of Shenzhen University (Luohu Hospital Group), Shenzhen, China
| | - Yueyue Zhang
- Department of Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Fan Li
- Department of Radiology, The Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, China
| | - Weihao Liu
- Shantou University Medical College, Shantou University, Shantou, China
| | - Haoyang Zeng
- Shantou University Medical College, Shantou University, Shantou, China
| | - Haoyuan Yuan
- Shantou University Medical College, Shantou University, Shantou, China
| | - Zixi Ye
- Shantou University Medical College, Shantou University, Shantou, China
| | - Zexin Huang
- Department of Radiology, Shenzhen Luohu District Traditional Chinese Medicine Hospital (Luohu Hospital Group), Shenzhen, China
| | - Yangguang Yuan
- Department of Radiology, The Third Affiliated Hospital of Shenzhen University (Luohu Hospital Group), Shenzhen, China
| | - Ye Xiang
- Department of Radiology, Leshan Hospital, Chengdu University of Traditional Chinese Medicine, Leshan, China.
| | - Kai Wu
- Department of Radiology, The Third Affiliated Hospital of Shenzhen University (Luohu Hospital Group), Shenzhen, China.
| | - Hanlin Liu
- Department of Radiology, The Third Affiliated Hospital of Shenzhen University (Luohu Hospital Group), Shenzhen, China.
| |
Collapse
|
10
|
Shin HB, Sheen H, Oh JH, Choi YE, Sung K, Kim HJ. Evaluating feature extraction reproducibility across image biomarker standardization initiative-compliant radiomics platforms using a digital phantom. J Appl Clin Med Phys 2025:e70110. [PMID: 40353843 DOI: 10.1002/acm2.70110] [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: 10/22/2024] [Revised: 03/27/2025] [Accepted: 04/07/2025] [Indexed: 05/14/2025] Open
Abstract
BACKGROUND The aim of this study was to thoroughly analyze the reproducibility of radiomics feature extraction across three Image Biomarker Standardization Initiative (IBSI)-compliant platforms using a digital phantom for benchmarking. It uncovers high consistency among common features while also pointing out the necessity for standardization in computational algorithms and mathematical definitions due to unique platform-specific features. METHODS We selected three widely used radiomics platforms: LIFEx, Computational Environment for Radiological Research (CERR), and PyRadiomics. Using the IBSI digital phantom, we performed a comparative analysis to extract and benchmark radiomics features. The study design included testing each platform's ability to consistently reproduce radiomics features, with statistical analyses to assess the variability and agreement among the platforms. RESULTS The results indicated varying levels of feature reproducibility across the platforms. Although some features showed high consistency, others varied significantly, highlighting the need for standardized computational algorithms. Specifically, LIFEx and PyRadiomics performed consistently well across many features, whereas CERR showed greater variability in certain feature categories than LIFEx and PyRadiomics. CONCLUSION The study findings highlight the need for harmonized feature calculation methods to enhance the reliability and clinical usefulness of radiomics. Additionally, this study recommends incorporating clinical data and establishing benchmarking procedures in future studies to enhance the role of radiomics in personalized medicine.
Collapse
Affiliation(s)
- Han-Back Shin
- Department of Radiation Oncology, Gachon University Gil Medical Center, Incheon, Republic of Korea
| | - Heesoon Sheen
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, Republic of Korea
- High-Energy Physics Center, Chung-Ang University, Seoul, Republic of Korea
| | - Jang-Hoon Oh
- Department of Radiology, Kyung Hee University Hospital, Kyung Hee University College of Medicine, Seoul, Republic of Korea
| | - Young Eun Choi
- Department of Radiation Oncology, Gachon University Gil Medical Center, Incheon, Republic of Korea
| | - Kihoon Sung
- Department of Radiation Oncology, Gil Medical Center, Gachon University College of Medicine, Incheon, Republic of Korea
| | - Hyun Ju Kim
- Department of Radiation Oncology, Gil Medical Center, Gachon University College of Medicine, Incheon, Republic of Korea
| |
Collapse
|
11
|
Mariotti F, Agostini A, Borgheresi A, Marchegiani M, Zannotti A, Giacomelli G, Pierpaoli L, Tola E, Galiffa E, Giovagnoni A. Insights into radiomics: a comprehensive review for beginners. Clin Transl Oncol 2025:10.1007/s12094-025-03939-5. [PMID: 40355777 DOI: 10.1007/s12094-025-03939-5] [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: 03/27/2025] [Accepted: 04/16/2025] [Indexed: 05/15/2025]
Abstract
Radiomics and artificial intelligence (AI) are rapidly evolving, significantly transforming the field of medical imaging. Despite their growing adoption, these technologies remain challenging to approach due to their technical complexity. This review serves as a practical guide for early-career radiologists and researchers seeking to integrate radiomics into their studies. It provides practical insights for clinical and research applications, addressing common challenges, limitations, and future directions in the field. This work offers a structured overview of the essential steps in the radiomics workflow, focusing on concrete aspects of each step, including indicative and practical examples. It covers the main steps such as dataset definition, image acquisition and preprocessing, segmentation, feature extraction and selection, and AI model training and validation. Different methods to be considered are discussed, accompanied by summary diagrams. This review equips readers with the knowledge necessary to approach radiomics and AI in medical imaging from a hands-on research perspective.
Collapse
Affiliation(s)
- Francesco Mariotti
- Department of Clinical, Special and Dental Sciences, University Politecnica delle Marche, Via Tronto, 10/A, 60126, Ancona, Italy
| | - Andrea Agostini
- Department of Clinical, Special and Dental Sciences, University Politecnica delle Marche, Via Tronto, 10/A, 60126, Ancona, Italy
- Department of Radiological Sciences - Division of Clinical Radiology, University Hospital "Azienda Ospedaliero Universitaria delle Marche", Via Conca, 71, 60126, Ancona, Italy
| | - Alessandra Borgheresi
- Department of Clinical, Special and Dental Sciences, University Politecnica delle Marche, Via Tronto, 10/A, 60126, Ancona, Italy.
- Department of Radiological Sciences - Division of Clinical Radiology, University Hospital "Azienda Ospedaliero Universitaria delle Marche", Via Conca, 71, 60126, Ancona, Italy.
| | - Marzia Marchegiani
- School of Radiology, University Politecnica delle Marche, Via Tronto, 10/A, 60126, Ancona, Italy
| | - Alice Zannotti
- School of Radiology, University Politecnica delle Marche, Via Tronto, 10/A, 60126, Ancona, Italy
| | - Gloria Giacomelli
- School of Radiology, University Politecnica delle Marche, Via Tronto, 10/A, 60126, Ancona, Italy
| | - Luca Pierpaoli
- School of Radiology, University Politecnica delle Marche, Via Tronto, 10/A, 60126, Ancona, Italy
| | - Elisabetta Tola
- School of Radiology, University Politecnica delle Marche, Via Tronto, 10/A, 60126, Ancona, Italy
| | - Elena Galiffa
- School of Radiology, University Politecnica delle Marche, Via Tronto, 10/A, 60126, Ancona, Italy
| | - Andrea Giovagnoni
- Department of Clinical, Special and Dental Sciences, University Politecnica delle Marche, Via Tronto, 10/A, 60126, Ancona, Italy
- Department of Radiological Sciences - Division of Clinical Radiology, University Hospital "Azienda Ospedaliero Universitaria delle Marche", Via Conca, 71, 60126, Ancona, Italy
| |
Collapse
|
12
|
Mali SA, Rad NM, Woodruff HC, Depeursinge A, Andrearczyk V, Lambin P. Harmonizing CT scanner acquisition variability in an anthropomorphic phantom: A comparative study of image-level and feature-level harmonization using GAN, ComBat, and their combination. PLoS One 2025; 20:e0322365. [PMID: 40344028 PMCID: PMC12063804 DOI: 10.1371/journal.pone.0322365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 03/20/2025] [Indexed: 05/11/2025] Open
Abstract
PURPOSE Radiomics allows for the quantification of medical images and facilitates precision medicine. Many radiomic features derived from computed tomography (CT) are sensitive to variations across scanners, reconstruction settings, and acquisition protocols. In this phantom study, eight different CT reconstruction parameters were varied to explore image- and feature-level harmonization approaches to improve tissue classification. METHODS Varying reconstructions of an anthropomorphic radiopaque phantom containing three lesion categories (metastasis, hemangioma, and benign cyst) and normal liver tissue were used for evaluating two harmonization methods and their combination: (i) generative adversarial networks (GANs) at the image level; (ii) ComBat at the feature level, and (iii) a combination of (i) and (ii). A total of 93 texture and intensity features were extracted from each tissue class before and after image-level harmonization and were also harmonized at the feature level. Reproducibility and stability were assessed via the Concordance Correlation Coefficient (CCC) and pairwise comparisons using paired stability tests. The ability of features to discriminate between tissue classes was assessed by measuring the area under the receiver operating characteristic curve. The global reproducibility and discriminative power were assessed by averaging over the entire dataset and across all tissue types. RESULTS ComBat improved reproducibility by 31.58% and stability by 5.24%, while GAN increased reproducibility by 8% it reduced stability by 4.33%. Classification analysis revealed that ComBat increased average AUC by 15.19%, whereas GAN decreased AUC by 2.56%. CONCLUSION While GAN qualitatively enhances image harmonization, ComBat provides superior statistical improvements in feature stability and classification performance, highlighting the importance of robust feature-level harmonization in radiomics.
Collapse
Affiliation(s)
- Shruti Atul Mali
- The D-Lab, Department of Precision Medicine, GROW- Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, Netherlands
| | - Nastaran Mohammadian Rad
- The D-Lab, Department of Precision Medicine, GROW- Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, Netherlands
| | - Henry C. Woodruff
- The D-Lab, Department of Precision Medicine, GROW- Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, Netherlands
- Department of Radiology and Nuclear Medicine, GROW- Research Institute School for Oncology and Reproduction, Maastricht University Medical Centre+, Maastricht, Netherlands
| | - Adrien Depeursinge
- Institute of Information Systems, University of Applied Sciences and Arts Western Switzerland, Sierre, Switzerland
- Department of Nuclear Medicine and Molecular Imaging, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Vincent Andrearczyk
- Institute of Information Systems, University of Applied Sciences and Arts Western Switzerland, Sierre, Switzerland
| | - Philippe Lambin
- The D-Lab, Department of Precision Medicine, GROW- Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, Netherlands
- Department of Radiology and Nuclear Medicine, GROW- Research Institute School for Oncology and Reproduction, Maastricht University Medical Centre+, Maastricht, Netherlands
| |
Collapse
|
13
|
Yang N, Ma ZX, Wang X, Xiao L, Jin L, Li M. Development and validation of a CT-based radiomics nomogram for predicting progression-free survival in patients with small cell lung cancer. BMC Med Imaging 2025; 25:154. [PMID: 40329257 PMCID: PMC12057258 DOI: 10.1186/s12880-025-01691-4] [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: 10/18/2024] [Accepted: 04/25/2025] [Indexed: 05/08/2025] Open
Abstract
PURPOSE Small cell lung cancer (SCLC) is a highly aggressive form of lung cancer, representing about 15% of cases worldwide. Despite advances in imaging, such as low-dose CT, which have increased diagnostic rates, survival outcomes for SCLC patients have remained stagnant. Recent studies have only focused on radiomics, which extracts detailed quantitative features from imaging, with clinical risk factors to improve prognostic models. Therefore, this study aimed to develop a clinical-radiomics fusion nomogram based on computed tomography (CT) to estimate progression-free survival (PFS) in patients diagnosed with SCLC. By integrating radiomics features extracted from CT with clinical data, this model provides personalized prognostic assessment for clinicians. Its clinical utility lies in aiding treatment decision-making by offering more accurate prognostic evaluation, optimizing therapeutic strategies, and identifying high-risk patients at an early stage, ultimately improving overall survival and quality of life. METHODS To develop the nomogram model, 95 patients diagnosed with pathologically confirmed SCLC between January 1, 2013, and December 31, 2023, were included in the study cohort. Participants were randomly divided into training and validation cohorts in a 7:3 ratio. Radiomics features associated with PFS were generated using the least absolute shrinkage and selection operator (LASSO) along with univariate and multivariate analyses. Additionally, in the training cohort, both univariate and multivariate analyses using Cox regression were conducted to identify the significant clinical risk factors influencing PFS. The predictive performance of the clinical and clinical-radiomics fusion nomogram were evaluated using the concordance index, calibration plots, and decision curve analysis (DCA). RESULTS Five radiomics features were selected and used to calculate the radiomics score (Rad-score). The radiomics features were significantly associated with PFS (hazard ratio: 0.5765, 95% confidence interval: 0.3641-0.9128, p < 0.05). Three clinical risk factors significantly associated with PFS were identified: neuron-specific enolase (NSE), carbohydrate antigen 125 levels (CA125), and treatment type, such as surgery. The clinical-radiomics fusion nomogram model (C-index:0.744) demonstrated superior performance compared to the clinical nomogram model (C-index: 0.718) in the training cohort. DCA indicated that the clinical-radiomics fusion nomogram outperformed the clinical nomogram in terms of clinical usefulness. CONCLUSIONS A CT-based clinical-radiomics fusion nomogram was developed to predict PFS in patients with SCLC, which is useful in providing individualized information. ADVANCES IN KNOWLEDGE A clinical-radiomics fusion nomogram was constructed to estimate the probability of PFS based on clinical risk factors and the rad-score.
Collapse
Affiliation(s)
- Nan Yang
- Department of Radiology, Huadong Hospital, Fudan University, Shanghai, 200040, China
- Zhang Guozhen Small Pulmonary Nodules Diagnosis and Treatment Center, Shanghai, 200040, China
| | - Zhuang Xuan Ma
- Department of Radiology, Huadong Hospital, Fudan University, Shanghai, 200040, China
- Zhang Guozhen Small Pulmonary Nodules Diagnosis and Treatment Center, Shanghai, 200040, China
| | - Xin Wang
- Department of Pathology, Huadong Hospital, Fudan University, Shanghai, 200040, China
| | - Li Xiao
- Department of Pathology, Huadong Hospital, Fudan University, Shanghai, 200040, China
| | - Liang Jin
- Department of Radiology, Huadong Hospital, Fudan University, Shanghai, 200040, China.
- Zhang Guozhen Small Pulmonary Nodules Diagnosis and Treatment Center, Shanghai, 200040, China.
| | - Ming Li
- Department of Radiology, Huadong Hospital, Fudan University, Shanghai, 200040, China.
- Zhang Guozhen Small Pulmonary Nodules Diagnosis and Treatment Center, Shanghai, 200040, China.
| |
Collapse
|
14
|
Al-Obeidat F, Rashid A, Hafez W, Gibbaoui H, Ayoub G, Al Ameer S, Venkatachalapathi AK, Gador M, Hassan S, Ibrahim MA, Hamza N, Cherrez-Ojeda I. The accuracy of artificial intelligence in the diagnosis of soft tissue sarcoma: A systematic review and meta-analysis. Curr Probl Surg 2025; 66:101743. [PMID: 40306879 DOI: 10.1016/j.cpsurg.2025.101743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 03/01/2025] [Accepted: 03/04/2025] [Indexed: 05/02/2025]
Affiliation(s)
- Feras Al-Obeidat
- College of Technological Innovation, Zayed University, Abu Dhabi Campus, Khalifa City, Abu Dhabi, UAE
| | - Asrar Rashid
- School of Computing, Edinburgh Napier University, Edinburgh, Scotland, UK.
| | - Wael Hafez
- NMC Royal Hospital, Abu Dhabi, UAE; Internal Medicine Department, Medical Research and Clinical Studies Institute, The National Research Centre, Cairo, Egypt
| | | | | | | | | | | | | | | | | | - Ivan Cherrez-Ojeda
- Universidad Espiritu Santo, Samborondon, Ecuador; Respiralab Research Group, Guayaquil, Ecuador
| |
Collapse
|
15
|
Hu Z, Xu M, Yang H, Hao H, Zhao P, Yang Y, Liu G. Development of an Intra- and Peritumoral Radiomics Nomogram Using Digital Breast Tomosynthesis for Preoperative Assessment of Ki-67 Expression in Invasive Breast Cancer. Acad Radiol 2025; 32:2465-2476. [PMID: 39915181 DOI: 10.1016/j.acra.2024.12.040] [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: 09/02/2024] [Revised: 11/10/2024] [Accepted: 12/18/2024] [Indexed: 04/23/2025]
Abstract
RATIONALE AND OBJECTIVES This study aimed to develop a radiomics nomogram model using preoperative digital breast tomosynthesis (DBT) images to predict Ki-67 expression in patients with invasive breast cancer (IBC). MATERIALS AND METHODS This retrospective study involved a cohort of 289 patients with IBC, who were randomly divided into a training dataset (N= 202) and a validation dataset (N= 87). Ki-67 expression was categorized into low and high groups using a 14% threshold. Radiomics features from both the intra- and peritumoral regions of DBT images were used to develop the radiomics model, referred to as Radscore. Clinical and nomogram models were constructed using multivariate logistic regression. The performance of the established models was evaluated using receiver operating characteristic (ROC) curve analysis, calibration curve analysis, decision curve analysis (DCA), net reclassification improvement (NRI), and integrated discrimination improvement (IDI). RESULTS The clinical model was constructed using tumor size and DBT-reported lymph node metastasis (DBT_reported_LNM). By integrating Radscore_Combine-which incorporates both intra- and peritumoral radiomics features-along with tumor size and DBT_reported_LNM into the nomogram, the model achieved the highest area under the curve (AUC) values of 0.819 and 0.755 in the training and validation datasets, respectively. The notable improvement shown by the NRI and IDI suggests that Radscore_Combine could serve as a valuable biomarker for predicting Ki-67 expression effectively. CONCLUSION The nomogram offers a non-invasive method to predict Ki-67 expression in IBC patients, which could aid in creating personalized treatment plans.
Collapse
Affiliation(s)
- Zhenzhen Hu
- Department of Radiology, China-Japan Union Hospital of Jilin University, Changchun, China (Z.H., H.H., Y.Y., G.L.)
| | - Maolin Xu
- Department of Radiology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (M.X.); Breast cancer center, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, National key clinical specialty construction discipline, Hubei Provincial Clinical Research Center for Breast Cancer, Wuhan Clinical Research Center for Breast Cancer, Wuhan, China (M.X.)
| | - Huimin Yang
- Department of Radiology, Linfen Central Hospital, Linfen, China (H.Y.)
| | - Haifeng Hao
- Department of Radiology, China-Japan Union Hospital of Jilin University, Changchun, China (Z.H., H.H., Y.Y., G.L.)
| | - Ping Zhao
- Department of Gastroenterology, China-Japan Union Hospital of Jilin University, Changchun, China (P.Z.)
| | - Yiqing Yang
- Department of Radiology, China-Japan Union Hospital of Jilin University, Changchun, China (Z.H., H.H., Y.Y., G.L.)
| | - Guifeng Liu
- Department of Radiology, China-Japan Union Hospital of Jilin University, Changchun, China (Z.H., H.H., Y.Y., G.L.).
| |
Collapse
|
16
|
Zhang W, Zhuang D, Wei W, Yang Y, Ma L, Du H, Jin A, He J, Li X. The 100 most-cited radiomics articles in cancer research: A bibliometric analysis. Clin Imaging 2025; 121:110442. [PMID: 40086035 DOI: 10.1016/j.clinimag.2025.110442] [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: 10/02/2024] [Revised: 02/15/2025] [Accepted: 03/06/2025] [Indexed: 03/16/2025]
Abstract
Radiomics, an advanced medical imaging analysis technique introduced by Professor Lambin in 2012, has quickly become a key area of medical research. To explore emerging trends in cancer-related radiomics, we conducted a bibliometric analysis of the 100 most-cited articles (T100) in this field. Data were collected from the Web of Science Core Collection on October 7, 2023, and the articles were ranked by citation count. We extracted data such as authors, journals, citation counts, and publication years and analyzed it using Microsoft Excel 2019 and R 4.4.2. CiteSpace was used to create co-occurrence and citation burst maps to show the relationships between authors, countries, institutions, and keywords. The analysis revealed that the T100 came from 81 countries, with the U.S. contributing the most (56 articles). Harvard University was the leading institution, and the journal Radiology had the highest citation count. Aerts Hugo JWL was the most influential author. The study highlights that "lung cancer" and "artificial intelligence" are emerging as major research hotspots, shaping the future of cancer radiomics.
Collapse
Affiliation(s)
- Wenhao Zhang
- Department of Psychiatry, Chaohu Hospital of Anhui Medical University, Hefei, Anhui, China; Department of Medical Psychology, School of Mental Health and Psychological Science, Anhui Medical University, Hefei, China; Department of Clinical Medical, First Clinical Medical College, Anhui Medical University, Hefei, China
| | - Dongmei Zhuang
- Suzhou Hospital of Anhui Medical University, Suzhou, Anhui, China
| | - Wenzhuo Wei
- Department of Medical Psychology, School of Mental Health and Psychological Science, Anhui Medical University, Hefei, China
| | - Yuchen Yang
- Department of Clinical Medical, First Clinical Medical College, Anhui Medical University, Hefei, China
| | - Lijun Ma
- Department of Medical Psychology, School of Mental Health and Psychological Science, Anhui Medical University, Hefei, China
| | - He Du
- Department of Medical Psychology, School of Mental Health and Psychological Science, Anhui Medical University, Hefei, China
| | - Anran Jin
- Department of Medical Psychology, School of Mental Health and Psychological Science, Anhui Medical University, Hefei, China
| | - Jingyi He
- Department of Medical Psychology, School of Mental Health and Psychological Science, Anhui Medical University, Hefei, China
| | - Xiaoming Li
- Department of Psychiatry, Chaohu Hospital of Anhui Medical University, Hefei, Anhui, China; Department of Medical Psychology, School of Mental Health and Psychological Science, Anhui Medical University, Hefei, China.
| |
Collapse
|
17
|
Bai G, Huo S, Wang G, Tian S. Artificial intelligence radiomics in the diagnosis, treatment, and prognosis of gynecological cancer: a literature review. Transl Cancer Res 2025; 14:2508-2532. [PMID: 40386259 PMCID: PMC12079260 DOI: 10.21037/tcr-2025-618] [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: 03/19/2025] [Accepted: 04/18/2025] [Indexed: 05/20/2025]
Abstract
Background and Objective Gynecological cancer is the most common cancer that affects women's quality of life and well-being. Artificial intelligence (AI) technology enables us to exploit high-dimensional imaging data for precision oncology. Tremendous progress has been made with AI radiomics in cancers such as lung and breast cancers. Herein, we performed a literature review on AI radiomics in the management of gynecological cancer. Methods A search was performed in the databases of PubMed, Embase, and Web of Science for original articles written in English up to 10 September 2024, using the terms "gynecological cancer", "cervical cancer", "endometrial cancer", "ovarian cancer", AND "artificial intelligence", "AI", AND "radiomics". The included studies mainly focused on the current landscape of AI radiomics in the diagnosis, treatment, and prognosis of gynecological cancer. Key Content and Findings A total of 128 studies were included, with 86 studies focusing on tumor diagnosis (n=23) and characterization (n=63), 15 on treatment response prediction, and 27 on recurrence and survival prediction. AI radiomics has shown potential value in tumor diagnosis and characterization [tumor staging, histological subtyping, lymph node metastasis (LNM), lymphovascular space invasion (LVSI), myometrial invasion (MI), and other molecular or clinicopathological factors], chemotherapy or chemoradiotherapy response evaluation, and prognosis (disease recurrence or metastasis, and survival) prediction. However, most included studies were single-center and retrospective. There was substantial heterogeneity in methodology and results reporting. Conclusions AI radiomics has been increasingly adopted in the management of gynecological cancer. Further validation in large-scale datasets is needed before clinical translation.
Collapse
Affiliation(s)
- Gengshen Bai
- Department of Intervention, The Second People’s Hospital of Baiyin City, Baiyin, China
| | - Shiwen Huo
- Jiangsu Hengrui Pharmaceuticals Co., Ltd., Shanghai, China
| | - Guangcai Wang
- Department of Intervention, The Second People’s Hospital of Baiyin City, Baiyin, China
| | - Shijia Tian
- Department of Intervention, The Second People’s Hospital of Baiyin City, Baiyin, China
| |
Collapse
|
18
|
Kaiser JD, Bräuherr F, Biesel EA, Chikhladze S, Fichtner-Feigl S, Ruess DA, Wittel UA. Preoperative prediction of postoperative pancreatic fistula after Pancreaticoduodenectomy: Determination and validation of a cut-off value for the Roberts Score. Am J Surg 2025; 245:116356. [PMID: 40319558 DOI: 10.1016/j.amjsurg.2025.116356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 04/15/2025] [Accepted: 04/17/2025] [Indexed: 05/07/2025]
Abstract
BACKGROUND POPF after pancreaticoduodenectomy can be life-threatening. For risk stratification, prediction could be key. The aim of this study is to determine and validate a cut-off value for the Roberts Score, which is one of the few purely preoperative multicenter validated predictive models for POPF. METHODS 582 patients were included. The Youden index determined a cut-off in the exploratory cohort (n = 466). The validation cohort's (n = 116) ability to predict CR-POPF was tested using univariate and multivariate regression analysis. RESULTS AUC of Roberts Score for the exploration cohort was 0.768. The identified cut-off of 0.268 was confirmed in the validation cohort (p < 0.001). Higher scores were significantly associated with longer time to drain removal and ICU stay. Multiple logistic regression showed the cut-off as an independent predictor of CR-POPF (p = 0.038). CONCLUSION The scoring variables and the cut-off itself were both independent predictors, which may improve the identification of high-risk patients and help to investigate the development of POPF.
Collapse
Affiliation(s)
- Johannes D Kaiser
- Department of General and Visceral Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany.
| | - Franziska Bräuherr
- Department of General and Visceral Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Esther A Biesel
- Department of General and Visceral Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Sophia Chikhladze
- Department of General and Visceral Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Stefan Fichtner-Feigl
- Department of General and Visceral Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Dietrich A Ruess
- Department of General and Visceral Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Uwe A Wittel
- Department of General and Visceral Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| |
Collapse
|
19
|
Chen Y, Guo W, Li Y, Lin H, Dong D, Qi Y, Pu R, Liu A, Li W, Sun B. Differentiation of Glioblastoma and Solitary Brain Metastasis Using Brain-Tumor Interface Radiomics Features Based on MR Images: A Multicenter Study. Acad Radiol 2025:S1076-6332(25)00308-3. [PMID: 40280830 DOI: 10.1016/j.acra.2025.04.008] [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: 02/10/2025] [Revised: 03/27/2025] [Accepted: 04/03/2025] [Indexed: 04/29/2025]
Abstract
RATIONALE AND OBJECTIVES Glioblastoma (GBM) and solitary brain metastasis (SBM) exhibit similar radiomics features on magnetic resonance imaging (MRI), yet their treatment strategies and prognoses significantly differ. Therefore, accurate differentiation between these two types of tumors is crucial for clinical decision-making. This study aims to establish and validate an efficient diagnostic model based on the radiomic features of the T1-weighted contrast-enhanced (T1CE) sequence in the 10 mm brain-tumor interface region to achieve precise differentiation between GBM and SBM. METHODS This study retrospectively collected contrast-enhanced T1-weighted imaging data from 226 GBM patients and 206 SBM patients at three centers between January 2010 and October 2024. Samples from centers 1 and 2 were used as the training set, while samples from center 3 were used as the test set. Two observers manually delineated the tumor edges on the T1CE images layer by layer to obtain the Region of Interest (ROI) covering the entire tumor volume. A 10 mm brain-to-tumor interface (BTI) was extracted using Python code. Radiomic features were extracted from the 10 mm BTI region, followed by feature selection and model construction. Finally, SHAP (SHapley Additive exPlanations) was used to visualize the model. Three radiologists with 2, 6, and 18 years of diagnostic experience independently evaluated the test set samples without knowing the patient information or pathology results, establishing three diagnostic models. The DeLong test was used to compare these models with the radiomic model. RESULTS Ultimately, ten radiomic features were used for modeling. The model established using the logistic regression (LR) algorithm had an AUC of 0.893 on the training set and 0.808 on the test set. The AUCs of the three radiologists with different diagnostic experiences on the test set were 0.699, 0.740, and 0.789, respectively, all lower than that of the radiomic model. The DeLong test showed that ModelBTI performed significantly better than Doctor 1 (p<0.05) in the test set, but there was no statistically significant difference in performance between ModelBTI and Doctors 2 and 3. CONCLUSION The radiomic model constructed based on the 10 mm brain-tumor interface can effectively differentiate between GBM and SBM, capturing tumor heterogeneity from a new perspective, thereby significantly improving diagnostic performance and providing assistance for clinical diagnosis. DATA AVAILABILITY STATEMENT The original contributions presented in the study are included in the article/Supplemental material, further inquiries can be directed to the corresponding authors.
Collapse
Affiliation(s)
- Yini Chen
- Department of Radiology, The First Affiliated Hospital of DalianMedical University, Dalian, China (Y.C., H.L., D.D., Y.Q., R.P., A.L., B.S.)
| | - Weiya Guo
- Department of Radiology, Dalian Municipal Women and Children's Medical Center (Group), Dalian, China (W.G.)
| | - Yushi Li
- Department of Radiology, The Second Affiliated Hospital of DalianMedical University, Dalian, China (Y.L.)
| | - Hongsen Lin
- Department of Radiology, The First Affiliated Hospital of DalianMedical University, Dalian, China (Y.C., H.L., D.D., Y.Q., R.P., A.L., B.S.)
| | - Deshuo Dong
- Department of Radiology, The First Affiliated Hospital of DalianMedical University, Dalian, China (Y.C., H.L., D.D., Y.Q., R.P., A.L., B.S.)
| | - Yiwei Qi
- Department of Radiology, The First Affiliated Hospital of DalianMedical University, Dalian, China (Y.C., H.L., D.D., Y.Q., R.P., A.L., B.S.)
| | - Renwang Pu
- Department of Radiology, The First Affiliated Hospital of DalianMedical University, Dalian, China (Y.C., H.L., D.D., Y.Q., R.P., A.L., B.S.)
| | - Ailian Liu
- Department of Radiology, The First Affiliated Hospital of DalianMedical University, Dalian, China (Y.C., H.L., D.D., Y.Q., R.P., A.L., B.S.)
| | - Wei Li
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China (W.L.)
| | - Bo Sun
- Department of Radiology, The First Affiliated Hospital of DalianMedical University, Dalian, China (Y.C., H.L., D.D., Y.Q., R.P., A.L., B.S.).
| |
Collapse
|
20
|
Shen Q, Xiang C, Han Y, Li Y, Huang K. The value of multi-phase CT based intratumor and peritumoral radiomics models for evaluating capsular characteristics of parotid pleomorphic adenoma. Front Med (Lausanne) 2025; 12:1566555. [PMID: 40330775 PMCID: PMC12054526 DOI: 10.3389/fmed.2025.1566555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Accepted: 04/03/2025] [Indexed: 05/08/2025] Open
Abstract
Objectives Computed tomography (CT) imaging of parotid pleomorphic adenoma (PA) has been widely reported, nonetheless few reports have estimated the capsule characteristics of PA at length. This study aimed to establish and validate CT-based intratumoral and peritumoral radiomics models to clarify the characteristics between parotid PA with and without complete capsule. Methods In total, data of 129 patients with PA were randomly assigned to a training and test set at a ratio of 7:3. Quantitative radiomics features of the intratumoral and peritumoral regions of 2 mm and 5 mm on CT images were extracted, and radiomics models of Tumor, External2, External5, Tumor+ External2, and Tumor+External5 were constructed and used to train six different machine learning algorithms. Meanwhile, the prediction performances of different radiomics models (Tumor, External2, External5, Tumor+External2, Tumor+External5) based on single phase (plain, arterial, and venous phase) and multiphase (three-phase combination) were compared. The receiver operating characteristic (ROC) curve analysis and the area under the curve (AUC) were used to evaluate the prediction performance of each model. Results Among all the established machine learning prediction radiomics models, the model based on a three-phase combination had better prediction performance, and the model using a combination of intratumoral and peritumoral radiomics features achieved a higher AUC than the model with only intratumoral or peritumoral radiomics features, and the Tumor+External2 model based on LR was the optimal model, the AUC of the test set was 0.817 (95% CI = 0.712, 0.847), and its prediction performance was significantly higher (p < 0.05, DeLong's test) than that with the Tumor model based on LDA (AUC of 0.772), the External2 model based on LR (AUC of 0.751), and the External5 model based on SVM (AUC of 0.667). And the Tumor+External2 model based on LR had a higher AUC than the Tumor+External5 model based on LDA (AUC = 0.817 vs. 0.796), but no statistically significant difference (P = 0.667). Conclusion The intratumoral and peritumoral radiomics model based on multiphasic CT images could accurately predict capsular characteristics of parotid of PA preoperatively, which may help in making treatment strategies before surgery, as well as avoid intraoperative tumor spillage and residuals.
Collapse
Affiliation(s)
- Qian Shen
- Department of Radiology, The Affiliated Stomatology Hospital of Southwest Medical University, Luzhou, China
| | - Cong Xiang
- School of Artificial Intelligence, Chongqing University of Technology, Chongqing, China
| | - Yongliang Han
- Department of Radiology, The First Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Yongmei Li
- Department of Radiology, The First Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Kui Huang
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatology Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
21
|
Liang X, Luo S, Liu Z, Liu Y, Luo S, Zhang K, Li L. Unsupervised machine learning analysis of optical coherence tomography radiomics features for predicting treatment outcomes in diabetic macular edema. Sci Rep 2025; 15:13389. [PMID: 40251316 PMCID: PMC12008428 DOI: 10.1038/s41598-025-96988-3] [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: 01/16/2025] [Accepted: 04/01/2025] [Indexed: 04/20/2025] Open
Abstract
This study aimed to identify distinct clusters of diabetic macular edema (DME) patients with differential anti-vascular endothelial growth factor (VEGF) treatment outcomes using an unsupervised machine learning (ML) approach based on radiomic features extracted from pre-treatment optical coherence tomography (OCT) images. Retrospective data from 234 eyes with DME treated with three anti-VEGF therapies between January 2020 and March 2024 were collected from two clinical centers. Radiomic analysis was conducted on pre-treatment OCT images. Following principal component analysis (PCA) for dimensionality reduction, two unsupervised clustering methods (K-means and hierarchical clustering) were applied. Baseline characteristics and treatment outcomes were compared across clusters to assess clustering efficacy. Feature selection employed a three-stage pipeline: exclusion of collinear features (Pearson's r > 0.8); sequential filtering through ANOVA (P < 0.05) and Boruta algorithm (500 iterations); multivariate stepwise regression (entry criteria: univariate P < 0.1) to identify outcome-associated predictors. From 1165 extracted radiomic features, four distinct DME clusters were identified. Cluster 4 exhibited a significantly lower incidence of residual/recurrent DME (RDME) (34.29%) compared to Clusters 1-3 (P = 0.003, P = 0.005 and P = 0.002, respectively). This cluster also demonstrated the highest proportion of eyes (71.43%) with best-corrected visual acuity (BCVA) exceeding 20/63 (P = 0.003, P = 0.005 and P = 0.002, respectively). Multivariate analysis identified logarithm_gldm_DependenceVariance as an independent risk factor for RDME (OR 1.75, 95% CI 1.28-2.40; P < 0.001), while Wavelet-LH_Firstorder_Mean correlated with worse visual outcomes (OR 8.76, 95% CI 1.22-62.84; P = 0.031). Unsupervised ML leveraging pre-treatment OCT radiomics successfully stratifies DME eyes into clinically distinct subgroups with divergent therapeutic responses. These quantitative features may serve as non-invasive biomarkers for personalized outcome prediction and retinal pathology assessment.
Collapse
Affiliation(s)
- Xuemei Liang
- Department of Ophthalmology, Aier Eye Hospital, Jinan University, No, 191, Huanshi Middle Road, Yuexiu District, Guangzhou, 510071, Guangdong, People's Republic of China
- Department of Ophthalmology, Nanning Aier Eye Hospital, No, 63, Chaoyang Road, Xingning District, Nanning, 530012, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Shaozhao Luo
- Department of Ophthalmology, Aier Eye Hospital, Jinan University, No, 191, Huanshi Middle Road, Yuexiu District, Guangzhou, 510071, Guangdong, People's Republic of China
| | - Zhigao Liu
- Department of Ophthalmology, Jinan Aier Eye Hospital, No. 1916, Erhuan East Road, Licheng District, Jinan City, Shandong Province, People's Republic of China
| | - Yunsheng Liu
- Department of Ophthalmology, Cenxi Aier Eye Hospital, No. 101, Yuwu Avenue, Cenxi City, Wuzhou City, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Shinan Luo
- Department of Ophthalmology, Nanning Aier Eye Hospital, No, 63, Chaoyang Road, Xingning District, Nanning, 530012, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Kaiqing Zhang
- Department of Ophthalmology, Aier Eye Hospital, Jinan University, No, 191, Huanshi Middle Road, Yuexiu District, Guangzhou, 510071, Guangdong, People's Republic of China
| | - Li Li
- Department of Ophthalmology, Aier Eye Hospital, Jinan University, No, 191, Huanshi Middle Road, Yuexiu District, Guangzhou, 510071, Guangdong, People's Republic of China.
- Department of Ophthalmology, Nanning Aier Eye Hospital, No, 63, Chaoyang Road, Xingning District, Nanning, 530012, Guangxi Zhuang Autonomous Region, People's Republic of China.
| |
Collapse
|
22
|
Zhang N, Ling H, Zhang W, Zhang M. A prediction method for radiation proctitis based on SAM-Med2D model. Sci Rep 2025; 15:13426. [PMID: 40251184 PMCID: PMC12008286 DOI: 10.1038/s41598-025-87409-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 01/20/2025] [Indexed: 04/20/2025] Open
Abstract
Cervical cancer, a prevalent gynecological malignancy, poses significant threats to women's health. Despite advances in treatment modalities, radiotherapy remains a cornerstone in managing cervical cancer. However, radiotherapy-induced complications, such as radiation proctitis, present substantial diagnostic and prognostic challenges. Accurate diagnosis are crucial for optimizing treatment strategies and improving patient outcomes. Deep learning has shown remarkable success in medical image segmentation, aiding clinicians in assessing patient conditions. In the other hand, radiomics excels in extracting diagnostically valuable features from medical images but requires extensive manual annotation and often lacks generalizability. Therefore, combining the strengths of deep learning and radiomics is pivotal in addressing these challenges. In this study, we propose a novel paradigm that leverages deep learning models for initial segmentation, followed by detailed radiomics analysis. Specifically, we utilize the Transformer-based SAM-Med2D model to extract visual features from CT images of cervical cancer patients. We apply T-tests and Lasso regression to identify features most correlated with radiation proctitis and build predictive models using logistic regression, random forest, and naive Gaussian Bayesian algorithms. Experimental results demonstrate that our method effectively extracts CT imaging features and exhibits excellent performance in diagnosis radiation proctitis. This approach not only enhances predictive accuracy but also provides a valuable tool for personalizing treatment plans and improving patient outcomes in cervical cancer radiotherapy.
Collapse
Affiliation(s)
- Ning Zhang
- Department of Radiotherapy, The First Affiliated Hospital of Anhui Medical University, Hefei, 230000, China
| | - Haifeng Ling
- School of Computer Science and Technology, University of Science and Technology of China, Hefei, 230000, China
| | - Wenyu Zhang
- School of Computer Science and Technology, University of Science and Technology of China, Hefei, 230000, China
| | - Mei Zhang
- Department of Radiotherapy, The First Affiliated Hospital of Anhui Medical University, Hefei, 230000, China.
- Oncology Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230000, China.
- Graduate School of Anhui University of Chinese Medicine, Hefei, 230000, China.
- The Traditional and Western Medicine (TCM)-Integrated Cancer Center of Anhui Medical University, Hefei, 230000, China.
| |
Collapse
|
23
|
Shen J, Zhang L, Li S, Mu X, Yu T, Zhang W, Yu Y, He J, Gao W. Multi-sequence MRI based radiomics nomogram for prediction expression of programmed death ligand 1 in thymic epithelial tumor. Front Immunol 2025; 16:1555530. [PMID: 40292290 PMCID: PMC12021882 DOI: 10.3389/fimmu.2025.1555530] [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: 01/04/2025] [Accepted: 03/20/2025] [Indexed: 04/30/2025] Open
Abstract
Background High expression levels of programmed death receptor 1 (PD-1) and its ligand 1 (PD-L1) have been observed in thymic epithelial tumors (TET), suggesting their potential as prognostic indicators for disease progression and the effectiveness of immunotherapy in TET. The conventional method obtaining PD-L1 was challenging due to invasive sampling and tumor heterogeneity. Methods A total of 124 patients with pathologically confirmed TET (57 PD-L1 positive, 67 PD-L1 negative) were retrospectively enrolled and allocated into training and validation cohorts in a ratio of 7:3. Radiomics features were extracted from T1-weighted, T2-weighted fat suppression, and apparent diffusion coefficient (ADC) map images to establish a radiomics signature in the training cohort. Multivariate logistic regression analysis was conducted to develop a combined radiomics nomogram that incorporated clinical, conventional MR features, or ADC model for evaluation purposes. The performance of each model was compared using receiver operating characteristics analysis, while discrimination, calibration, and clinical efficiency of the combined radiomics nomogram were assessed. Results The radiomics signature, consisting of four features, demonstrated a favorable ability to predict and differentiate between PD-L1 positive and negative TET patients. The combined radiomics nomogram, which incorporates the peri-cardial invasion sign, ADC value, WHO classification, and radiomics signature, showed excellent performance (training cohort: area under the curve [AUC] = 0.903; validation cohorts: AUC = 0.894). The calibration curve and decision curve analysis further confirmed the clinical usefulness of this combined model. The decision curve analysis demonstrated the clinical utility of the integrated radiomics nomogram. Conclusions The radiomics signature serves as a valuable tool for predicting the PD-L1 status of TET patients. Furthermore, the integration of radiomics nomogram enhances the personalized prediction capability.
Collapse
Affiliation(s)
- Jie Shen
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lantian Zhang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Shuke Li
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaofei Mu
- Department of Oncology, The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yining, China
| | - Tongfu Yu
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wei Zhang
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yue Yu
- Department of Thoracic Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jing He
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wen Gao
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
24
|
Wang G, Wang S, Song W, Lu C, Chen Z, He L, Wang X, Wang Y, Shi C, Liu Z, Yu Y, Wang X, Tian Y, Li Y. Integrating multi-omics data reveals the antitumor role and clinical benefits of gamma-delta T cells in triple-negative breast cancer. BMC Cancer 2025; 25:623. [PMID: 40197136 PMCID: PMC11974128 DOI: 10.1186/s12885-025-14029-8] [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: 12/09/2024] [Accepted: 03/26/2025] [Indexed: 04/09/2025] Open
Abstract
BACKGROUND Gamma-delta (γδ) T cells are a critical component of the tumor microenvironment and have been recognized as a promising biomarker and target for cancer therapy. Increasing evidence suggests that γδT cells play distinct roles in different cancers. However, the impact of γδT cells in breast cancer remains controversial. METHODS In this study, we investigated the role of γδT cells in breast cancer using a comprehensive approach, including bulk and single-cell sequencing, radiomics based on magnetic resonance imaging (MRI), genomic data, and immunohistochemistry. Single-cell RNA profiling was used to infer the potential lineage evolution of γδT cells and their interactions with other immune cells. Bulk RNA sequencing was included to uncover the heterogeneity in signaling pathways, as well as radiotherapy and immunotherapy responses, among patients with varying levels of γδT cell abundance. Genomic analysis was used to recognize the critical gene mutations with the infiltration of γδT cells. Immunohistochemistry was performed to validate the prognostic value of γδT cells in breast cancer patients. Lastly, radiomics was used to establish a correlation between the abundance of γδT cells and the features of MRI images. RESULTS The γδT cell infiltration was closely associated with favorable prognosis in triple-negative breast cancer (TNBC) but not in other subtypes of breast cancer. γδT cells may exert antitumor effects through intrinsic lineage evolution or interact with antigen-presenting cells through ligand-receptor pairs. Patients with a high γδT cell abundance may benefit more from chemotherapy or radiotherapy alone than their combination. Additionally, patients with a high γδT cell abundance were more likely to benefit from immunotherapy. Finally, we established a radiomic model based on dynamic contrast-enhanced-MRI, which indicated the potential for estimating the γδT cell abundance for patients with TNBC. CONCLUSION Our study provides novel insight and a theoretical basis for individualized therapy of patients with TNBC based on γδT cells.
Collapse
Affiliation(s)
- Guixin Wang
- The First Department of Breast Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Huan-Hu-Xi Road, He-Xi District, Tianjin, 300060, China
- Immunology Department, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Medical University, Qixiangtai Road 22, He-Ping District, Tianjin, 300070, China
| | - Shuo Wang
- The First Department of Breast Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Huan-Hu-Xi Road, He-Xi District, Tianjin, 300060, China
| | - Wenbin Song
- Department of General Surgery, Tianjin Key Laboratory of Precise Vascular Reconstruction and Organ Function Repair, Tianjin Medical University General Hospital, Tianjin General Surgery Institute, An-Shan Road 154, He-Ping District, Tianjin, 300052, China
| | - Chenglu Lu
- Department of Pathology, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin Cancer Institute and Hospital, Tianjin Medical University, Tianjin, 300060, China
- Department of Pathology, Tangshan People's Hospital, Tangshan, Hebei, 063001, China
| | - Zhaohui Chen
- The First Department of Breast Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Huan-Hu-Xi Road, He-Xi District, Tianjin, 300060, China
| | - Long He
- Department of General Surgery, Tianjin Key Laboratory of Precise Vascular Reconstruction and Organ Function Repair, Tianjin Medical University General Hospital, Tianjin General Surgery Institute, An-Shan Road 154, He-Ping District, Tianjin, 300052, China
| | - Xiaoning Wang
- Department of General Surgery, Tianjin Key Laboratory of Precise Vascular Reconstruction and Organ Function Repair, Tianjin Medical University General Hospital, Tianjin General Surgery Institute, An-Shan Road 154, He-Ping District, Tianjin, 300052, China
| | - Yizeng Wang
- Department of General Surgery, Tianjin Key Laboratory of Precise Vascular Reconstruction and Organ Function Repair, Tianjin Medical University General Hospital, Tianjin General Surgery Institute, An-Shan Road 154, He-Ping District, Tianjin, 300052, China
| | - Cangchang Shi
- Department of General Surgery, Tianjin Key Laboratory of Precise Vascular Reconstruction and Organ Function Repair, Tianjin Medical University General Hospital, Tianjin General Surgery Institute, An-Shan Road 154, He-Ping District, Tianjin, 300052, China
| | - Zhaoyi Liu
- Department of General Surgery, Tianjin Key Laboratory of Precise Vascular Reconstruction and Organ Function Repair, Tianjin Medical University General Hospital, Tianjin General Surgery Institute, An-Shan Road 154, He-Ping District, Tianjin, 300052, China
| | - Yue Yu
- The First Department of Breast Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Huan-Hu-Xi Road, He-Xi District, Tianjin, 300060, China
| | - Xin Wang
- The First Department of Breast Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Huan-Hu-Xi Road, He-Xi District, Tianjin, 300060, China.
| | - Yao Tian
- The First Department of Breast Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Huan-Hu-Xi Road, He-Xi District, Tianjin, 300060, China.
- Department of General Surgery, Tianjin Key Laboratory of Precise Vascular Reconstruction and Organ Function Repair, Tianjin Medical University General Hospital, Tianjin General Surgery Institute, An-Shan Road 154, He-Ping District, Tianjin, 300052, China.
| | - Yingxi Li
- Immunology Department, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Medical University, Qixiangtai Road 22, He-Ping District, Tianjin, 300070, China.
| |
Collapse
|
25
|
Yang R, Shi Z, Ruan J, Li Z, Li Y, You R, Liu L, Li W, Chen X. Application of peritumoral radiomics based on simulated positioning CT images in the prognosis of intermediate-advanced esophageal cancer. Sci Rep 2025; 15:11865. [PMID: 40195320 PMCID: PMC11977252 DOI: 10.1038/s41598-024-82392-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 12/05/2024] [Indexed: 04/09/2025] Open
Abstract
This study aimed to develop a prognostic model utilizing intratumoral and peritumoral radiomics from simulated localization CT images to predict overall survival (OS) in patients with advanced esophageal cancer, while evaluating its clinical applicability. We conducted a retrospective cohort study involving 151 patients with esophageal cancer who underwent radical radiotherapy between January 2017 and January 2023 (144 men, 7 women). Participants were randomly assigned to a training cohort (n = 105) and a validation cohort (n = 46) at a 7:3 ratio. The primary outcome measured was OS. We extracted 851 radiomic features from the radiotherapy target area of localized CT images. Univariate Cox and LASSO-Cox models were employed to identify features associated with OS. We developed four Cox proportional hazards regression models: a clinical model, a GTV radiomics model combined with the clinical model, a peritumoral radiomics model combined with the clinical model, and a comprehensive radiomics-clinical model. Model performance was assessed using receiver operating characteristic (ROC) curves, Kaplan-Meier survival curves, and nomograms. The median follow-up period was 22 months (range: 6-101). The clinical model exhibited C-index values of 0.540 and 0.590 for predicting OS in the training and validation cohorts, respectively. The GTV radiomics combined with the clinical model demonstrated improved performance with C-index values of 0.753 and 0.677. The peritumoral radiomics combined with the clinical model yielded C-index values of 0.662 and 0.587. The total radiomics-clinical model showed the best predictive capability, with C-index values of 0.762 and 0.704 in the training and validation cohorts. Calibration curves validated the accuracy and clinical relevance of the total radiomics-clinical model, which effectively stratified patient risk categories (p < 0.001). The total radiomics-clinical model, developed from simulated localization CT images, demonstrates a robust ability to predict overall survival (OS) in patients with advanced esophageal cancer. By accurately identifying high- and low-risk patients, this model empowers clinicians to tailor treatment strategies to individual patient profiles. This personalized approach enhances clinical decision-making, enabling more effective allocation of resources and interventions based on the unique prognostic factors of each patient.
Collapse
Affiliation(s)
- Ruiling Yang
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Centre, Kunming, 650118, China
| | - Zhihui Shi
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Centre, Kunming, 650118, China
| | - Jinqiu Ruan
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Centre, Kunming, 650118, China
| | - Zhenhui Li
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Centre, Kunming, 650118, China
| | - Yanli Li
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Centre, Kunming, 650118, China
| | - Ruimin You
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Centre, Kunming, 650118, China
| | - Lizhu Liu
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Centre, Kunming, 650118, China
| | - Wang Li
- Department of Thoracic surgery, The Third Affiliated Hospital of Kunming Medical University, Kunming, China.
- Department of Thoracic surgery, Yunnan Cancer Hospital, Kunming, China.
| | - Xiaobo Chen
- Department of Radiation Therapy, The Third Affiliated Hospital of Kunming Medical University, Kunming, China.
- Department of Radiation Therapy, Yunnan Cancer Hospital, Kunming, China.
| |
Collapse
|
26
|
Du S, Xie W, Gao S, Zhao R, Wang H, Tian J, Liu J, Liu Z, Zhang L. Early prediction of neoadjuvant therapy response in breast cancer using MRI-based neural networks: data from the ACRIN 6698 trial and a prospective Chinese cohort. Breast Cancer Res 2025; 27:52. [PMID: 40181457 PMCID: PMC11969705 DOI: 10.1186/s13058-025-02009-6] [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: 12/08/2024] [Accepted: 03/24/2025] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND Early prediction of treatment response to neoadjuvant therapy (NAT) in breast cancer patients can facilitate timely adjustment of treatment regimens. We aimed to develop and validate a MRI-based enhanced self-attention network (MESN) for predicting pathological complete response (pCR) based on longitudinal images at the early stage of NAT. METHODS Two imaging datasets were utilized: a subset from the ACRIN 6698 trial (dataset A, n = 227) and a prospective collection from a Chinese hospital (dataset B, n = 245). These datasets were divided into three cohorts: an ACRIN 6698 training cohort (n = 153) from dataset A, an ACRIN 6698 test cohort (n = 74) from dataset A, and an external test cohort (n = 245) from dataset B. The proposed MESN allowed for the integration of multiple timepoint features and extraction of dynamic information from longitudinal MR images before and after early-NAT. We also constructed the Pre model based on pre-NAT MRI features. Clinicopathological characteristics were added to these image-based models to create integrated models (MESN-C and Pre-C), and their performance was evaluated and compared. RESULTS The MESN-C yielded area under the receiver operating characteristic curve (AUC) values of 0.944 (95% CI: 0.906 - 0.973), 0.903 (95%CI: 0.815 - 0.965), and 0.861 (95%CI: 0.811 - 0.906) in the ACRIN 6698 training, ACRIN 6698 test and external test cohorts, respectively, which were significantly higher than those of the clinical model (AUC: 0.720 [95%CI: 0.587 - 0.842], 0.738 [95%CI: 0.669 - 0.796] for the two test cohorts, respectively; p < 0.05) and Pre-C (AUC: 0.697 [95%CI: 0.554 - 0.819], 0.726 [95%CI: 0.666 - 0.797] for the two test cohorts, respectively; p < 0.05). High AUCs of the MESN-C maintained in the ACRIN 6698 standard (AUC = 0.853 [95%CI: 0.676 - 1.000]) and experimental (AUC = 0.905 [95%CI: 0.817 - 0.993]) subcohorts, and the interracial and external subcohort (AUC = 0.861 [95%CI: 0.811 - 0.906]). Moreover, the MESN-C increased the positive predictive value from 48.6 to 71.3% compared with Pre-C model, and maintained a high negative predictive value (80.4-86.7%). CONCLUSION The MESN-C using longitudinal multiparametric MRI after a short-term therapy achieved favorable performance for predicting pCR, which could facilitate timely adjustment of treatment regimens, increasing the rates of pCR and avoiding toxic effects. TRIAL REGISTRATION Trial registration at https://www.chictr.org.cn/ . REGISTRATION NUMBER ChiCTR2000038578, registered September 24, 2020.
Collapse
Affiliation(s)
- Siyao Du
- Department of Radiology, The First Hospital of China Medical University, Shenyang, 110001, Liaoning Province, China
| | - Wanfang Xie
- School of Engineering Medicine, Beihang University, Beijing, 100191, People's Republic of China
- Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology of the People's Republic of China, Beijing, 100191, People's Republic of China
| | - Si Gao
- Department of Radiology, The First Hospital of China Medical University, Shenyang, 110001, Liaoning Province, China
| | - Ruimeng Zhao
- Department of Radiology, The First Hospital of China Medical University, Shenyang, 110001, Liaoning Province, China
| | - Huidong Wang
- Department of Radiology, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110165, Liaoning Province, China
| | - Jie Tian
- School of Engineering Medicine, Beihang University, Beijing, 100191, People's Republic of China.
- Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology of the People's Republic of China, Beijing, 100191, People's Republic of China.
| | - Jiangang Liu
- School of Engineering Medicine, Beihang University, Beijing, 100191, People's Republic of China.
- Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology of the People's Republic of China, Beijing, 100191, People's Republic of China.
- Beijing Engineering Research Center of Cardiovascular Wisdom Diagnosis and Treatment, Beijing, 100029, People's Republic of China.
| | - Zhenyu Liu
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Beijing, 100190, People's Republic of China.
- University of Chinese Academy of Sciences, Beijing, 100080, People's Republic of China.
| | - Lina Zhang
- Department of Radiology, The First Hospital of China Medical University, Shenyang, 110001, Liaoning Province, China.
- Department of Radiology, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110165, Liaoning Province, China.
| |
Collapse
|
27
|
Lo Mastro A, Grassi E, Berritto D, Russo A, Reginelli A, Guerra E, Grassi F, Boccia F. Artificial intelligence in fracture detection on radiographs: a literature review. Jpn J Radiol 2025; 43:551-585. [PMID: 39538068 DOI: 10.1007/s11604-024-01702-4] [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: 07/26/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Fractures are one of the most common reasons of admission to emergency department affecting individuals of all ages and regions worldwide that can be misdiagnosed during radiologic examination. Accurate and timely diagnosis of fracture is crucial for patients, and artificial intelligence that uses algorithms to imitate human intelligence to aid or enhance human performs is a promising solution to address this issue. In the last few years, numerous commercially available algorithms have been developed to enhance radiology practice and a large number of studies apply artificial intelligence to fracture detection. Recent contributions in literature have described numerous advantages showing how artificial intelligence performs better than doctors who have less experience in interpreting musculoskeletal X-rays, and assisting radiologists increases diagnostic accuracy and sensitivity, improves efficiency, and reduces interpretation time. Furthermore, algorithms perform better when they are trained with big data on a wide range of fracture patterns and variants and can provide standardized fracture identification across different radiologist, thanks to the structured report. In this review article, we discuss the use of artificial intelligence in fracture identification and its benefits and disadvantages. We also discuss its current potential impact on the field of radiology and radiomics.
Collapse
Affiliation(s)
- Antonio Lo Mastro
- Department of Radiology, University of Campania "Luigi Vanvitelli", Naples, Italy.
| | - Enrico Grassi
- Department of Orthopaedics, University of Florence, Florence, Italy
| | - Daniela Berritto
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Anna Russo
- Department of Radiology, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Alfonso Reginelli
- Department of Radiology, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Egidio Guerra
- Emergency Radiology Department, "Policlinico Riuniti Di Foggia", Foggia, Italy
| | - Francesca Grassi
- Department of Radiology, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Francesco Boccia
- Department of Radiology, University of Campania "Luigi Vanvitelli", Naples, Italy
| |
Collapse
|
28
|
Li M, Zheng A, Song M, Jin F, Pang M, Zhang Y, Wu Y, Li X, Zhao M, Li Z. From text to insight: A natural language processing-based analysis of burst and research trends in HER2-low breast cancer patients. Ageing Res Rev 2025; 106:102692. [PMID: 39993452 DOI: 10.1016/j.arr.2025.102692] [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: 03/15/2024] [Revised: 01/01/2025] [Accepted: 02/10/2025] [Indexed: 02/26/2025]
Abstract
With the intensification of population aging, the proportion of elderly breast cancer patients is continuously increasing, among which those with low HER2 expression account for approximately 45 %-55 % of all cases within traditional HER2-negative breast cancer. Concurrently, the significant therapeutic effect of T-DXd on patients with HER2-low tumors has brought this group into the public spotlight. Since the clinical approval of T-DXd in 2019, there has been a significant vertical surge in the volume of publications within this domain. We analyzed 512 articles on HER2-low breast cancer from the Web of Science Core Collection using bibliometrics, topic modeling, and knowledge graph techniques to summarize the current state and trends of research in this domain. Research efforts are particularly concentrated in the United States and China. Our analysis revealed six main research directions: HER2 detection, omics and clinical biomarkers, basic and translational research, neoadjuvant therapy and prognosis, progress of ADC drugs and clinical trials. To enhance the therapeutic efficacy and safety of antibodydrug conjugates (ADCs), researchers are actively exploring potential drug candidates other than T-DXd, with numerous ADC drugs emerging in clinical practice and trials. By incorporating emerging treatment strategies such as immunotherapy and employing circulating tumor cell (CTC) detection techniques, progress has been made toward improving the prognosis of patients with low HER2 expression. We believe that these research efforts hold promise as compelling evidence that HER2-low breast cancer may constitute a distinct and independent subtype.
Collapse
Affiliation(s)
- Muyao Li
- Department of Breast Surgery, the First Hospital of China Medical University, Shenyang, Liaoning 110001, China.
| | - Ang Zheng
- Department of Breast Surgery, the First Hospital of China Medical University, Shenyang, Liaoning 110001, China.
| | - Mingjie Song
- Department of General Medicine, the First Hospital of China Medical University, Shenyang, Liaoning 110001, China.
| | - Feng Jin
- Department of Breast Surgery, the First Hospital of China Medical University, Shenyang, Liaoning 110001, China.
| | - Mengyang Pang
- Department of Breast Surgery, the First Hospital of China Medical University, Shenyang, Liaoning 110001, China.
| | - Yuchong Zhang
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang, Liaoning 110001, China.
| | - Ying Wu
- Department of General Medicine, the First Hospital of China Medical University, Shenyang, Liaoning 110001, China; Phase I Clinical Trails Center, The First Hospital of China Medical University, Shenyang, Liaoning 110101, China.
| | - Xin Li
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang, Liaoning 110001, China.
| | - Mingfang Zhao
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang, Liaoning 110001, China.
| | - Zhi Li
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang, Liaoning 110001, China; National Clinical Research Center for Laboratory Medicine, Department of Laboratory Medicine, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China; Research Unit of Medical Laboratory, Chinese Academy of Medical Sciences, China.
| |
Collapse
|
29
|
Li Y, Yang L, Gu X, Wang X, Wang Q, Shi G, Zhang A, Deng H, Zhao X, Ren J, Miao A, Li S. Radiomics to predict PNI in ESCC. Abdom Radiol (NY) 2025; 50:1475-1487. [PMID: 39311949 PMCID: PMC11947035 DOI: 10.1007/s00261-024-04562-8] [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: 06/30/2024] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 03/27/2025]
Abstract
OBJECTIVE This study aimed to investigate whether contrast-enhanced computed tomography (CECT) based radiomics analysis could noninvasively predict the perineural invasion (PNI) in esophageal squamous cell carcinoma (ESCC). METHODS 398 patients with ESCC who underwent resection between February 2016 and March 2020 were retrospectively enrolled in this study. Patients were randomly divided into training and testing cohorts in a 7:3 ratio. Radiomics analysis was performed on the arterial phase images of CECT scans. From these images, 1595 radiomics features were initially extracted. The intraclass correlation coefficient (ICC), wilcoxon rank-sum test, spearman correlation analysis, and boruta algorithm were used for feature selection. Logistic regression (LR), random forest (RF), and support vector machine (SVM) models were established to preidict the PNI status. The performance of these radiomics models was assessed by the area under the receiver operating characteristic curve (AUC). Decision curve analysis (DCA) was conducted to evaluate their clinical utility. RESULTS Six radiomics features were retained to build the radiomics models. Among these models, the random forest (RF) model demonstrated superior performance. In the training cohort, the AUC value of the RF model was 0.773, compared to 0.627 for the logistic regression (LR) model and 0.712 for the support vector machine (SVM) model. Similarly, in the testing cohort, the RF model achieved an AUC value of 0.767, outperforming the LR model at 0.638 and the SVM model at 0.683. Decision curve analysis (DCA) suggested that the RF radiomics model exhibited the highest clinical utility. CONCLUSIONS CECT-based radiomics analysis, particularly utilizing the RF, can noninvasively predict the PNI in ESCC preoperatively. This novel approach could enhance patient management by providing personalized information, thereby facilitating the development of individualized treatment strategies for ESCC patients.
Collapse
Affiliation(s)
- Yang Li
- The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Li Yang
- The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiaolong Gu
- The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiangming Wang
- The Fourth Hospital of Hebei Medical University, Shijiazhuang, China.
| | - Qi Wang
- The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Gaofeng Shi
- The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Andu Zhang
- The Fourth Hospital of Hebei Medical University, Shijiazhuang, China.
| | - Huiyan Deng
- The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiaopeng Zhao
- The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | | | - Aijun Miao
- The Fourth People's Hospital of Hengshui, Hengshui, China
| | - Shaolian Li
- The Fourth People's Hospital of Hengshui, Hengshui, China
| |
Collapse
|
30
|
Li Y, Deng J, Ma X, Li W, Wang Z. Diagnostic accuracy of CT and PET/CT radiomics in predicting lymph node metastasis in non-small cell lung cancer. Eur Radiol 2025; 35:1966-1979. [PMID: 39223336 DOI: 10.1007/s00330-024-11036-4] [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: 04/18/2024] [Revised: 06/09/2024] [Accepted: 08/07/2024] [Indexed: 09/04/2024]
Abstract
OBJECTIVES This study evaluates the accuracy of radiomics in predicting lymph node metastasis in non-small cell lung cancer, which is crucial for patient management and prognosis. METHODS Adhering to PRISMA and AMSTAR guidelines, we systematically reviewed literature from March 2012 to December 2023 using databases including PubMed, Web of Science, and Embase. Radiomics studies utilizing computed tomography (CT) and positron emission tomography (PET)/CT imaging were included. The quality of studies was appraised with QUADAS-2 and RQS tools, and the TRIPOD checklist assessed model transparency. Sensitivity, specificity, and AUC values were synthesized to determine diagnostic performance, with subgroup and sensitivity analyses probing heterogeneity and a Fagan plot evaluating clinical applicability. RESULTS Our analysis incorporated 42 cohorts from 22 studies. CT-based radiomics demonstrated a sensitivity of 0.84 (95% CI: 0.79-0.88, p < 0.01) and specificity of 0.82 (95% CI: 0.75-0.87, p < 0.01), with an AUC of 0.90 (95% CI: 0.87-0.92), indicating no publication bias (p-value = 0.54 > 0.05). PET/CT radiomics showed a sensitivity of 0.82 (95% CI: 0.76-0.86, p < 0.01) and specificity of 0.86 (95% CI: 0.81-0.90, p < 0.01), with an AUC of 0.90 (95% CI: 0.87-0.93), with a slight publication bias (p-value = 0.03 < 0.05). Despite high clinical utility, subgroup analysis did not clarify heterogeneity sources, suggesting influences from possible factors like lymph node location and small subgroup sizes. CONCLUSIONS Radiomics models show accuracy in predicting lung cancer lymph node metastasis, yet further validation with larger, multi-center studies is necessary. CLINICAL RELEVANCE STATEMENT Radiomics models using CT and PET/CT imaging may improve the prediction of lung cancer lymph node metastasis, aiding personalized treatment strategies. RESEARCH REGISTRATION UNIQUE IDENTIFYING NUMBER (UIN) International Prospective Register of Systematic Reviews (PROSPERO), CRD42023494701. This study has been registered on the PROSPERO platform with a registration date of 18 December 2023. https://www.crd.york.ac.uk/prospero/ KEY POINTS: The study explores radiomics for lung cancer lymph node metastasis detection, impacting surgery and prognosis. Radiomics improves the accuracy of lymph node metastasis prediction in lung cancer. Radiomics can aid in the prediction of lymph node metastasis in lung cancer and personalized treatment.
Collapse
Affiliation(s)
- Yuepeng Li
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, China
| | - Junyue Deng
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Xuelei Ma
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| | - Weimin Li
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, China
- Institute of Respiratory Health, West China Hospital, Sichuan University, Chengdu, China
- Precision Medicine Center, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
- The Research Units of West China, Chinese Academy of Medical Sciences, West China Hospital, Chengdu, China
| | - Zhoufeng Wang
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, China.
- Institute of Respiratory Health, West China Hospital, Sichuan University, Chengdu, China.
- Precision Medicine Center, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China.
- The Research Units of West China, Chinese Academy of Medical Sciences, West China Hospital, Chengdu, China.
| |
Collapse
|
31
|
Huang X, Qin M, Fang M, Wang Z, Hu C, Zhao T, Qin Z, Zhu H, Wu L, Yu G, De Cobelli F, Xie X, Palumbo D, Tian J, Dong D. The application of artificial intelligence in upper gastrointestinal cancers. JOURNAL OF THE NATIONAL CANCER CENTER 2025; 5:113-131. [PMID: 40265096 PMCID: PMC12010392 DOI: 10.1016/j.jncc.2024.12.006] [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: 06/13/2024] [Revised: 09/17/2024] [Accepted: 12/20/2024] [Indexed: 04/24/2025] Open
Abstract
Upper gastrointestinal cancers, mainly comprising esophageal and gastric cancers, are among the most prevalent cancers worldwide. There are many new cases of upper gastrointestinal cancers annually, and the survival rate tends to be low. Therefore, timely screening, precise diagnosis, appropriate treatment strategies, and effective prognosis are crucial for patients with upper gastrointestinal cancers. In recent years, an increasing number of studies suggest that artificial intelligence (AI) technology can effectively address clinical tasks related to upper gastrointestinal cancers. These studies mainly focus on four aspects: screening, diagnosis, treatment, and prognosis. In this review, we focus on the application of AI technology in clinical tasks related to upper gastrointestinal cancers. Firstly, the basic application pipelines of radiomics and deep learning in medical image analysis were introduced. Furthermore, we separately reviewed the application of AI technology in the aforementioned aspects for both esophageal and gastric cancers. Finally, the current limitations and challenges faced in the field of upper gastrointestinal cancers were summarized, and explorations were conducted on the selection of AI algorithms in various scenarios, the popularization of early screening, the clinical applications of AI, and large multimodal models.
Collapse
Affiliation(s)
- Xiaoying Huang
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
| | - Minghao Qin
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
- University of Science and Technology Beijing, Beijing, China
| | - Mengjie Fang
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University, Beijing, China
- Key Laboratory of Big Data-Based Precision Medicine, Beihang University, Ministry of Industry and Information Technology, Beijing, China
| | - Zipei Wang
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
| | - Chaoen Hu
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
| | - Tongyu Zhao
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
- University of Science and Technology of China, Hefei, China
| | - Zhuyuan Qin
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- Beijing University of Chinese Medicine, Beijing, China
| | | | - Ling Wu
- KiangWu Hospital, Macau, China
| | | | | | | | - Diego Palumbo
- Department of Radiology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University, Beijing, China
- Key Laboratory of Big Data-Based Precision Medicine, Beihang University, Ministry of Industry and Information Technology, Beijing, China
| | - Di Dong
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
32
|
Wang Y, Hu Z, Wang H. The clinical implications and interpretability of computational medical imaging (radiomics) in brain tumors. Insights Imaging 2025; 16:77. [PMID: 40159380 PMCID: PMC11955438 DOI: 10.1186/s13244-025-01950-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 03/06/2025] [Indexed: 04/02/2025] Open
Abstract
Radiomics has widespread applications in the field of brain tumor research. However, radiomic analyses often function as a 'black box' due to their use of complex algorithms, which hinders the translation of brain tumor radiomics into clinical applications. In this review, we will elaborate extensively on the application of radiomics in brain tumors. Additionally, we will address the interpretability of handcrafted-feature radiomics and deep learning-based radiomics by integrating biological domain knowledge of brain tumors with interpretability methods. Furthermore, we will discuss the current challenges and prospects concerning the interpretability of brain tumor radiomics. Enhancing the interpretability of radiomics may make it more understandable for physicians, ultimately facilitating its translation into clinical practice. CRITICAL RELEVANCE STATEMENT: The interpretability of brain tumor radiomics empowers neuro-oncologists to make well-informed decisions from radiomic models. KEY POINTS: Radiomics makes a significant impact on the management of brain tumors in several key clinical areas. Transparent models, habitat analysis, and feature attribute explanations can enhance the interpretability of traditional handcrafted-feature radiomics in brain tumors. Various interpretability methods have been applied to explain deep learning-based models; however, there is a lack of biological mechanisms underlying these models.
Collapse
Affiliation(s)
- Yixin Wang
- Department of Brain Oncology, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, P. R. China
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, P. R. China
| | - Zongtao Hu
- Department of Brain Oncology, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, P. R. China
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, P. R. China
| | - Hongzhi Wang
- Department of Brain Oncology, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, P. R. China.
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, P. R. China.
| |
Collapse
|
33
|
Yang Z, Wang S, Yin W, Wang Y, Liu F, Xu J, Han L, Liu C. Radiomics-clinical nomogram for preoperative tumor-node-metastasis staging prediction in breast cancer patients using dynamic enhanced magnetic resonance imaging. Transl Cancer Res 2025; 14:1836-1848. [PMID: 40225004 PMCID: PMC11985186 DOI: 10.21037/tcr-24-1559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 01/09/2025] [Indexed: 04/15/2025]
Abstract
Background Breast cancer is one of the most commonly diagnosed malignancies in women worldwide, and the disease burden continues to aggravate. The tumor-node-metastasis (TNM) staging information is crucial for oncology physicians to develop appropriate clinical strategies. This study aimed to investigate the value of a radiomics-clinical model for predicting TNM stage in breast cancer patients using dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI). Methods DCE-MRI images from 166 patients with pathologically confirmed breast cancer were retrospectively collected, including early stage (TNM0-TNM2) and locally advanced or advanced stage (TNM3-TNM4). Included patients were divided into a training cohort (n=116) and a test cohort (n=50). The radiomics, clinical and integrated models were constructed and a nomogram was established to distinguish the TNM0-TNM2 stage from the TNM3-TNM4 stage. Receiver operating characteristic (ROC) curves, calibration curves and decision curve analysis (DCA) were employed to assess the predictability of the models. Results Eighty-five patients were at the early stages, while 81 patients were at the other stages. In the training and test cohorts, the area under the curve (AUC) values for distinguishing early and advanced breast cancer were 0.870 and 0.818 for the nomogram, respectively. The nomogram calibration curves showed good agreement between the predicted and observed TNM stages in the training and test cohorts. The Hosmer-Lemeshow test showed that the nomogram fit perfectly in the two cohorts. DCA indicated that the nomogram displayed clear superiority in forecasting TNM staging over clinical and radiomic signatures. Conclusions Compared to traditional imaging methods, the clinical-radiomics nomogram acquired by DCE-MRI could potentially be utilized to preoperatively evaluate the TNM stage of breast cancer with relatively high accuracy. It can be an effective method to guide clinical decisions.
Collapse
Affiliation(s)
- Zhe Yang
- Department of Radiology, the Second Affiliated Hospital of Shandong First Medical University, Tai’an, China
| | - Shouen Wang
- Department of Pathology, the Second Affiliated Hospital of Shandong First Medical University, Tai’an, China
| | - Wei Yin
- Department of Radiology, Beijing Friendship Hospital of Capital Medical University, Beijing, China
| | - Ying Wang
- Department of Radiology, the First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Fanghua Liu
- Department of Radiology, the Second Affiliated Hospital of Shandong First Medical University, Tai’an, China
| | - Jianshu Xu
- Department of Radiology, the Second Affiliated Hospital of Shandong First Medical University, Tai’an, China
| | - Long Han
- Department of Radiology, the Second Affiliated Hospital of Shandong First Medical University, Tai’an, China
| | - Chenglong Liu
- Department of Radiology, the Second Affiliated Hospital of Shandong First Medical University, Tai’an, China
| |
Collapse
|
34
|
He J, Liu N, Zhao L. New progress in imaging diagnosis and immunotherapy of breast cancer. Front Immunol 2025; 16:1560257. [PMID: 40165974 PMCID: PMC11955504 DOI: 10.3389/fimmu.2025.1560257] [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: 01/14/2025] [Accepted: 03/03/2025] [Indexed: 04/02/2025] Open
Abstract
Breast cancer (BC) is a predominant malignancy among women globally, with its etiology remaining largely elusive. Diagnosis primarily relies on invasive histopathological methods, which are often limited by sample representation and processing time. Consequently, non-invasive imaging techniques such as mammography, ultrasound, and Magnetic Resonance Imaging (MRI) are indispensable for BC screening, diagnosis, staging, and treatment monitoring. Recent advancements in imaging technologies and artificial intelligence-driven radiomics have enhanced precision medicine by enabling early detection, accurate molecular subtyping, and personalized therapeutic strategies. Despite reductions in mortality through traditional treatments, challenges like tumor heterogeneity and therapeutic resistance persist. Immunotherapies, particularly PD-1/PD-L1 inhibitors, have emerged as promising alternatives. This review explores recent developments in BC imaging diagnostics and immunotherapeutic approaches, aiming to inform clinical practices and optimize therapeutic outcomes.
Collapse
Affiliation(s)
- Jie He
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Nan Liu
- Department of Translational Medicine and Clinical Research, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Li Zhao
- Department of Radiology, Shaoxing People’s Hospital, Shaoxing, Zhejiang, China
| |
Collapse
|
35
|
Wang Y, Wan X, Liu Z, Liu Z, Huang X. Radiomics-based prediction of recurrent acute pancreatitis in individuals with metabolic syndrome using T2WI magnetic resonance imaging data. Front Med (Lausanne) 2025; 12:1502315. [PMID: 40115788 PMCID: PMC11922943 DOI: 10.3389/fmed.2025.1502315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 02/24/2025] [Indexed: 03/23/2025] Open
Abstract
Objective This study sought to clarify the utility of T2-weighted imaging (T2WI)-based radiomics to predict the recurrence of acute pancreatitis (AP) in subjects with metabolic syndrome (MetS). Methods Data from 196 patients with both AP and MetS from our hospital were retrospectively analyzed. These patients were separated into two groups according to their clinical follow-up outcomes, including those with first-onset AP (n = 114) and those with recurrent AP (RAP) (n = 82). The 196 cases were randomly divided into a training set (n = 137) and a test set (n = 59) at a 7:3 ratio. The clinical characteristics of these patients were systematically compiled for further analysis. For each case, the pancreatic parenchyma was manually delineated slice by slice using 3D Slicer software, and the appropriate radiomics characteristics were retrieved. The K-best approach, the least absolute shrinkage and selection operator (LASSO) algorithm, and variance thresholding were all used in the feature selection process. The establishment of clinical, radiomics, and combined models for forecasting AP recurrence in patients with MetS was then done using a random forest classifier. Model performance was measured using the area under the receiver operating characteristic curve (AUC), and model comparison was done using the DeLong test. The clinical utility of these models was evaluated using decision curve analysis (DCA), and the optimal model was determined via a calibration curve. Results In the training set, the clinical, radiomics, and combined models yielded respective AUCs of 0.651, 0.825, and 0.883, with corresponding test sets of AUCs of 0.606, 0.776, and 0.878. Both the radiomics and combined models exhibited superior predictive effectiveness compared to the clinical model in both the training (p = 0.001, p < 0.001) and test sets (p = 0.04, p < 0.001). The combined model outperformed the radiomics model (training set: p = 0.025, test set: p = 0.019). The DCA demonstrated that the radiomics and combined models had greater clinical efficacy than the clinical model. The calibration curve for the combined model demonstrated good agreement between the predicted probability of AP recurrence and the observed outcomes. Conclusion These findings highlight the superior predictive power of a T2WI-based radiomics model for predicting AP recurrence in patients with MetS, potentially supporting early interventions that can mitigate or alleviate RAP.
Collapse
Affiliation(s)
- Yuan Wang
- Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Xiyao Wan
- Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Ziyan Liu
- Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Ziyi Liu
- Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Xiaohua Huang
- Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| |
Collapse
|
36
|
Yang SX, Li M, Zhou LN, Hou DH, Zhang L, Wu N. Reproducibility of the CT radiomic features of pulmonary nodules: the effects of the CT reconstruction algorithm, radiation dose, and contrast agent. Quant Imaging Med Surg 2025; 15:2309-2318. [PMID: 40160618 PMCID: PMC11948441 DOI: 10.21037/qims-24-2026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 01/14/2025] [Indexed: 04/02/2025]
Abstract
Background The reproducibility of radiomic features (RFs) is essential in lung nodule diagnosis. This study aimed to prospectively investigate the effects of computed tomography (CT) scanning parameters on the reproducibility of RFs in pulmonary nodules. Methods Patients with pulmonary nodules who underwent chest CT scans at the Cancer Hospital of the Chinese Academy of Medical Sciences between July 2018 and March 2019 were prospectively included in the study. Six sequences with three pairs of different scanning parameters, including the reconstruction algorithm [filtered back projection (FBP) vs. 50% adaptive statistical iterative reconstruction-V (ASiR-V)], radiation dose (low dose vs. standard dose), and contrast agent [contrast-enhanced (CE) CT vs. non-contrast enhanced (NE) CT], were used for each patient. When one of the scanning parameters was changed, the other two remained fixed. The nodules were classified into pure ground-glass nodules (pGGNs), part-solid nodules (PSNs), and solid nodules (SNs) according to the nodule consistency. RFs with an intraclass correlation coefficient (ICC) >0.75 were considered to have good retest reliability. All the RF values of the different scanning parameters and nodule consistency were investigated and compared. Results A total of 150 pulmonary nodules, including 50 pGGNs, 50 PSNs, and 50 SNs, in 96 patients (mean age: 52±10 years; 62 females) were included in the study. In total, 320 RFs with an ICC >0.75 were evaluated. The proportion of RFs showed significant difference between FBP and 50% ASiR-V, low dose and standard dose, and CE and NE CT scans was 38.4% (123/320), 63.1% (202/320) and 54.1% (173/320), respectively. The radiation dose and contrast agent affected more RFs than the reconstruction algorithm (both P<0.001). In the subgroup analysis of nodule consistency, regardless of changes in the reconstruction algorithms, radiation doses, or contrast agents, the RFs showed significant difference among the pGGNs, PSNs, and SNs (all P<0.001). Conclusions The scanning parameters affected the reproducibility of the RFs, and nodules of different consistency were affected differently. The effects of these parameters should be fully considered in radiomic analysis.
Collapse
Affiliation(s)
- Shou-Xin Yang
- 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
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Radiology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Meng Li
- 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
| | - Li-Na Zhou
- 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
| | - Dong-Hui Hou
- 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
| | - Li Zhang
- 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
| | - Ning Wu
- 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
- Department of Nuclear Medicine (PET-CT Center), National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
37
|
Wu L, Zhu Y, Huang Q, Chen S, Zhou H, Xu Z, Li B, Chen H, Lv J. Research on imaging biomarkers for chronic subdural hematoma recurrence. Med Biol Eng Comput 2025; 63:823-834. [PMID: 39500853 DOI: 10.1007/s11517-024-03232-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 10/22/2024] [Indexed: 03/11/2025]
Abstract
This study utilizes radiomics to explore imaging biomarkers for predicting the recurrence of chronic subdural hematoma (CSDH), aiming to improve the prediction of CSDH recurrence risk. Analyzing CT scans from 64 patients with CSDH, we extracted 107 radiomic features and employed recursive feature elimination (RFE) and the XGBoost algorithm for feature selection and model construction. The feature selection process identified six key imaging biomarkers closely associated with CSDH recurrence: flatness, surface area to volume ratio, energy, run entropy, small area emphasis, and maximum axial diameter. The selection of these imaging biomarkers was based on their significance in predicting CSDH recurrence, revealing deep connections between postoperative variables and recurrence. After feature selection, there was a significant improvement in model performance. The XGBoost model demonstrated the best classification performance, with the average accuracy improving from 46.82% (before feature selection) to 80.74% and the AUC value increasing from 0.5864 to 0.7998. These results prove that precise feature selection significantly enhances the model's predictive capability. This study not only reveals imaging biomarkers for CSDH recurrence but also provides valuable insights for future personalized treatment strategies.
Collapse
Affiliation(s)
- Liyang Wu
- School of Life & Environmental Science, Guilin University of Electronic Technology, Guilin, 541004, China
| | - Yvmei Zhu
- School of Life & Environmental Science, Guilin University of Electronic Technology, Guilin, 541004, China
| | - Qiuyong Huang
- School of Life & Environmental Science, Guilin University of Electronic Technology, Guilin, 541004, China
| | - Shuchao Chen
- School of Life & Environmental Science, Guilin University of Electronic Technology, Guilin, 541004, China
| | - Haoyang Zhou
- School of Life & Environmental Science, Guilin University of Electronic Technology, Guilin, 541004, China
| | - Zihao Xu
- Department of Neurosurgery, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Bo Li
- Department of Neurosurgery, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Hongbo Chen
- School of Life & Environmental Science, Guilin University of Electronic Technology, Guilin, 541004, China.
- Guangxi Human Physiological Information Non-Invasive Detection Engineering Technology Research Center, Guilin, 541004, China.
- Guangxi Colleges and Universities Key Laboratory of Biomedical Sensors and Intelligent Instruments, Guilin, 541004, China.
- Guangxi Key Laboratory of Metabolic Reprogramming and Intelligent Medical Engineering for Chronic Diseases, Guilin, 541004, China.
| | - Junhui Lv
- Department of Neurosurgery, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, 310016, China.
| |
Collapse
|
38
|
Huang X, Cao Y, Zhang G, Tang F, Sun D, Ren J, Li W, Zhou J, Zhang J. MRI morphological features combined with apparent diffusion coefficient can predict brain invasion in meningioma. Comput Biol Med 2025; 187:109763. [PMID: 39908915 DOI: 10.1016/j.compbiomed.2025.109763] [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: 12/30/2023] [Revised: 01/12/2025] [Accepted: 01/28/2025] [Indexed: 02/07/2025]
Abstract
OBJECTIVES Accurately predicting meningioma brain invasion preoperatively helps to select the appropriate surgical approach and predict prognosis, but there are few imaging features that are sufficient for discriminating it alone. We investigate the joint MR imaging features and apparent diffusion coefficient (ADC) to predict the risk of brain invasion of meningiomas preoperatively. METHODS In this retrospective study, 143 patients (invasion group:51, non-invasion group: 92) diagnosed with meningioma by histopathology were included. The maximum (ADCmax), minimum (ADCmin) and mean (ADCmean) values of ADC and the mean ADC values of a comparative ROI in the normal appearing white matter (ADCNAWM) were calculated. Differences between clinical features, MRI morphological features, and all ADC values were assessed by Pearson's chi-square test and Kruskal-Wallis rank-sum test. Stepwise logistic regression analysis was used to select the optimal features and construct a prediction model. Furthermore, A nomogram was used to predict the risk of brain invasion, and a decision curve was used to verify the clinical utility of the nomogram. RESULTS According to stepwise logistic regression analysis, we found that sex, maximum diameter, peritumoral edema and ADCmin were closely related to brain invasion in meningioma. The model of the above four variables has the optimal discriminative ability to predict brain invasion, with an AUC of 0.924 (95 % CI, 0.879-0.969) and a sensitivity of 92.2 % (95 % CI, 74.5%-98.0 %). CONCLUSIONS Combining clinical features, MRI morphological characteristics and ADCmin, the model exhibits excellent discriminatory ability and high sensitivity, which can be used for predicting the risk of brain invasion of meningiomas.
Collapse
Affiliation(s)
- Xiaoyu Huang
- Department of Radiology, The Fifth Affiliated Hospital of Zunyi Medical University, Zhuhai, China
| | - Yuntai Cao
- Department of Radiology, Affiliated Hospital of Qinghai University, Xining, China
| | - Guojin Zhang
- Department of Radiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - FuQiang Tang
- Department of Nursing, Zhuhai Campus of Zunyi Medical University, Zhuhai, China
| | - Dandan Sun
- Department of Radiology, The Fifth Affiliated Hospital of Zunyi Medical University, Zhuhai, China
| | - Jialiang Ren
- Shanghai United Imaging Research Institute of Intelligent Imaging, Shanghai, China
| | - Wenyi Li
- Department of Radiology, The Fifth Affiliated Hospital of Zunyi Medical University, Zhuhai, China
| | - Junlin Zhou
- Department of Radiology, The Second Hospital of Lanzhou University, Lanzhou, China
| | - Jing Zhang
- Department of Radiology, The Fifth Affiliated Hospital of Zunyi Medical University, Zhuhai, China.
| |
Collapse
|
39
|
Shi L, Li C, Bai Y, Cao Y, Zhao S, Chen X, Cheng Z, Zhang Y, Li H. CT radiomics to predict pathologic complete response after neoadjuvant immunotherapy plus chemoradiotherapy in locally advanced esophageal squamous cell carcinoma. Eur Radiol 2025; 35:1594-1604. [PMID: 39470794 DOI: 10.1007/s00330-024-11141-4] [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: 03/10/2024] [Revised: 08/26/2024] [Accepted: 09/19/2024] [Indexed: 11/01/2024]
Abstract
OBJECTIVE To develop and validate a CT-based radiomics model to predict pathologic complete response (pCR) after neoadjuvant immunotherapy plus chemoradiotherapy (NICRT) in locally advanced esophageal squamous cell carcinoma (ESCC). METHODS A total of 105 patients with locally advanced ESCC receiving NICRT from February 2019 to December 2023 were enrolled. Patients were randomly divided into the training cohort and the test cohort at a 3:1 ratio. Enhanced CT scans were obtained before NICRT treatment. The 2D and 3D regions of interest were segmented, and features were extracted, followed by feature selection. Six algorithms were applied to construct the radiomics and clinical models. These models were evaluated by area under curve (AUC), accuracy, sensitivity, and specificity, and their respective optimal algorithms were further compared. RESULTS Forty-eight patients (45.75%) achieved pCR after NICRT. The AUC values of three algorithms in 2D radiomics models were higher than those in the 3D radiomics model and clinical model. Among these, the 2D radiomics model based on eXtreme Gradient Boosting (XGBoost) exhibited the best performance, with an AUC of 0.89 (95% CI, 0.81-0.97), accuracy of 0.85, sensitivity of 0.86, and specificity of 0.84 in the training cohort, and an AUC of 0.80 (95% CI, 0.64-0.97), accuracy of 0.77, sensitivity of 0.84, and specificity of 0.69 in the test cohort. Calibration curves also showed good agreement between predicted and actual response, and the decision curve analysis further confirmed its clinical applicability. CONCLUSION The 2D radiomics model can effectively predict pCR to NICRT in locally advanced ESCC. KEY POINTS Question Can CT-based radiomics predict pathologic complete response (pCR) after neoadjuvant immunotherapy plus chemoradiotherapy (NICRT) in locally advanced esophageal squamous cell carcinoma (ESCC)? Findings The model based on eXtreme Gradient Boosting (XGBoost) performed best, with an AUC of 0.89 in the training and 0.80 in the test cohort. Clinical relevance This CT-based radiomics model exhibits promising performance for predicting pCR to NICRT in locally advanced ESCC, which may be valuable in personalized treatment plan optimization.
Collapse
Affiliation(s)
- Liqiang Shi
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Chengqiang Li
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yaya Bai
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yuqin Cao
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Shengguang Zhao
- Department of Radiotherapy, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiaoyan Chen
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Zenghui Cheng
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yajie Zhang
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Hecheng Li
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
40
|
Liang ZY, Yu ML, Yang H, Li HJ, Xie H, Cui CY, Zhang WJ, Luo C, Cai PQ, Lin XF, Liu KF, Xiong L, Liu LZ, Chen BY. Beyond the tumor region: Peritumoral radiomics enhances prognostic accuracy in locally advanced rectal cancer. World J Gastroenterol 2025; 31:99036. [PMID: 40062323 PMCID: PMC11886509 DOI: 10.3748/wjg.v31.i8.99036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 10/09/2024] [Accepted: 11/05/2024] [Indexed: 01/23/2025] Open
Abstract
BACKGROUND The peritumoral region possesses attributes that promote cancer growth and progression. However, the potential prognostic biomarkers in this region remain relatively underexplored in radiomics. AIM To investigate the prognostic value and importance of peritumoral radiomics in locally advanced rectal cancer (LARC). METHODS This retrospective study included 409 patients with biopsy-confirmed LARC treated with neoadjuvant chemoradiotherapy and surgically. Patients were divided into training (n = 273) and validation (n = 136) sets. Based on intratumoral and peritumoral radiomic features extracted from pretreatment axial high-resolution small-field-of-view T2-weighted images, multivariate Cox models for progression-free survival (PFS) prediction were developed with or without clinicoradiological features and evaluated with Harrell's concordance index (C-index), calibration curve, and decision curve analyses. Risk stratification, Kaplan-Meier analysis, and permutation feature importance analysis were performed. RESULTS The comprehensive integrated clinical-radiological-omics model (ModelICRO) integrating seven peritumoral, three intratumoral, and four clinicoradiological features achieved the highest C-indices (0.836 and 0.801 in the training and validation sets, respectively). This model showed robust calibration and better clinical net benefits, effectively distinguished high-risk from low-risk patients (PFS: 97.2% vs 67.6% and 95.4% vs 64.8% in the training and validation sets, respectively; both P < 0.001). Three most influential predictors in the comprehensive ModelICRO were, in order, a peritumoral, an intratumoral, and a clinicoradiological feature. Notably, the peritumoral model outperformed the intratumoral model (C-index: 0.754 vs 0.670; P = 0.015); peritumoral features significantly enhanced the performance of models based on clinicoradiological or intratumoral features or their combinations. CONCLUSION Peritumoral radiomics holds greater prognostic value than intratumoral radiomics for predicting PFS in LARC. The comprehensive model may serve as a reliable tool for better stratification and management postoperatively.
Collapse
Affiliation(s)
- Zhi-Ying Liang
- Department of Radiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
| | - Mao-Li Yu
- Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
- West China School of Medicine, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Hui Yang
- Department of Radiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
| | - Hao-Jiang Li
- Department of Radiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
| | - Hui Xie
- Department of Radiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
| | - Chun-Yan Cui
- Department of Radiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
| | - Wei-Jing Zhang
- Department of Radiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
| | - Chao Luo
- Department of Radiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
| | - Pei-Qiang Cai
- Department of Radiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
| | - Xiao-Feng Lin
- Department of Radiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
| | - Kun-Feng Liu
- Department of Radiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
| | - Lang Xiong
- Department of Medical Imaging, First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, Jiangxi Province, China
| | - Li-Zhi Liu
- Department of Radiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
| | - Bi-Yun Chen
- Department of Radiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
| |
Collapse
|
41
|
Yu Y, Li GF, Tan WX, Qu XY, Zhang T, Hou XY, Zhu YB, Ma ZY, Yang L, Gao Y, Yu M, Yue C, Zhou Z, Yang Y, Yan LF, Cui GB. Towards automatical tumor segmentation in radiomics: a comparative analysis of various methods and radiologists for both region extraction and downstream diagnosis. BMC Med Imaging 2025; 25:63. [PMID: 40000987 PMCID: PMC11863488 DOI: 10.1186/s12880-025-01596-2] [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: 10/07/2024] [Accepted: 02/14/2025] [Indexed: 02/27/2025] Open
Abstract
OBJECTIVE By discussing the difference, stability and classification ability of tumor contour extracted by artificial intelligence and doctors, can a more stable method of tumor contour extraction be obtained? METHODS We propose a novel framework for the automatic segmentation of lung tumor contours and the differential diagnosis of downstream tasks. This framework integrates four key modules: tumor segmentation, extraction of radiomic features, feature selection, and the development of diagnostic models for clinical applications. Using this framework, we conducted a study involving a cohort of 1,429 patients suspected of lung cancer. Four automatic segmentation methods (RNN, UNET, WFCM, and SNAKE) were evaluated against manual segmentation performed by three radiologists with varying levels of expertise. We further studied the consistency of radiomic features extracted from these methods and evaluates their diagnostic performance across three downstream tasks: benign vs. malignant classification, lung adenocarcinoma infiltration, and lung nodule density classification. RESULTS The Dice coefficient of RNN is the highest among the four automatic segmentation methods (0.803 > 0.751, 0.576, 0.560), and all P < 0.05. In the consistency comparison of the seven contour-extracted radiomic features, that the features extracted by RNN and S1 (the senior radiologist) showed the highest similarity which was higher than the other automatic segmentation methods and doctors with low seniority. In all three downstream tasks, the radiomic features extracted from RNN segmentation contours showed the highest diagnostic discrimination. In the classification of benign and malignant nodules, the RNN method performed slightly better than the S1 method, with an AUC of 0.840 ± 0.01 and 0.824 ± 0.015, respectively, and significantly better than the other five methods. Similarly, the RNN method had an AUC value of 0.946 in lung adenocarcinoma infiltration, and a kappa value of 0.729 in lung nodule density classification, both of which were better than the other six methods. CONCLUSIONS Our findings suggest that AI-driven tumor segmentation methods can enhance clinical decision-making by providing reliable and reproducible results, ultimately emphasizing the auxiliary role of automated tumor contouring in clinical practice. The findings will have important implications for the application of radiomics in clinical practice.
Collapse
Affiliation(s)
- Ying Yu
- Department of Radiology & Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), 569 Xinsi Road, Xi'an , Shaanxi, 710038, China
| | - Gang-Feng Li
- Department of Radiology & Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), 569 Xinsi Road, Xi'an , Shaanxi, 710038, China
| | - Wei-Xiong Tan
- Deepwise Artificial Intelligence (AI), Deepwise Inc, 8 Haidian Street, Beijing, 100080, China
| | - Xiao-Yan Qu
- Department of Radiology & Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), 569 Xinsi Road, Xi'an , Shaanxi, 710038, China
| | - Tao Zhang
- Department of Pulmonary and Critical Care Medicine, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), 569 Xinsi Road, Xi'an, Shaanxi, 710038, China
| | - Xing-Yi Hou
- Department of Radiology & Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), 569 Xinsi Road, Xi'an , Shaanxi, 710038, China
| | - Yuan-Bo Zhu
- Department of Radiology & Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), 569 Xinsi Road, Xi'an , Shaanxi, 710038, China
| | - Zhi-Ying Ma
- Department of Radiology & Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), 569 Xinsi Road, Xi'an , Shaanxi, 710038, China
| | - Lu Yang
- Department of Radiology & Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), 569 Xinsi Road, Xi'an , Shaanxi, 710038, China
| | - Ya Gao
- Department of Radiology & Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), 569 Xinsi Road, Xi'an , Shaanxi, 710038, China
| | - Mei Yu
- Department of Radiology & Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), 569 Xinsi Road, Xi'an , Shaanxi, 710038, China
| | - Cui Yue
- Department of Radiology & Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), 569 Xinsi Road, Xi'an , Shaanxi, 710038, China
| | - Zhen Zhou
- Deepwise Artificial Intelligence (AI), Deepwise Inc, 8 Haidian Street, Beijing, 100080, China
| | - Yang Yang
- Department of Radiology & Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), 569 Xinsi Road, Xi'an , Shaanxi, 710038, China.
| | - Lin-Feng Yan
- Department of Radiology & Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), 569 Xinsi Road, Xi'an , Shaanxi, 710038, China.
| | - Guang-Bin Cui
- Department of Radiology & Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), 569 Xinsi Road, Xi'an , Shaanxi, 710038, China.
| |
Collapse
|
42
|
Sun K, Chen G, Liu C, Chu Z, Huang L, Li Z, Zhong S, Ye X, Zhang Y, Jia Y, Pan J, Zhou G, Liu Z, Yu C, Wang Y. A novel MSN-II feature extracted from T1-weighted MRI for discriminating between BD patients and MDD patients. J Affect Disord 2025; 371:36-44. [PMID: 39557301 DOI: 10.1016/j.jad.2024.11.047] [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: 05/24/2024] [Revised: 10/16/2024] [Accepted: 11/15/2024] [Indexed: 11/20/2024]
Abstract
BACKGROUND Differentiating between patients with bipolar disorder (BD) and major depressive disorder (MDD) is clinically challenging. This study aimed to explore the potential of radiomic textural features for discriminating BD and MDD. METHODS A total 253 subjects (114 patients with BD, 139 patients with MDD) with T1-weighted MRI data were recruited. Radiomics features and gray matter volume (GMV) features were extracted from each brain region. A novel high-level MSN_II feature method based on radiomic features was proposed. And a total of 21 MSN features (5 MSN_I and 16 MSN_II) based on different combinations of the 5 types of radiomic textural feature were calculated. Classification models were constructed using various combinations of MSNs or GMV, and their performance and stability was evaluated through 2000 repeated experiments. RESULTS The model built with combined features (GMV and GMV + MSN_II_GLCM_GLSZM_NGTDM) showed the best classification performance (AUC = 0.896±0.058, ACC = 0.831±0.064) in the validation cohort. After MANOVA analysis and FDR correlation, the MSN_II_GLCM_GLSZM_NGTDM values in 4 regions (right rectus gyrus, right temporal pole: middle temporal gyrus, Vermis3 and Vermis10) showed significant difference between BD and MDD. LIMITATION The main limitation of this study is that the data is derived from a single center without an external independent test set. CONCLUSIONS Incorporating the high-level MSN_II based on radiomics features can improve the classification performance compared to models solely relying on GMV features alone. This result implied the potential application of the proposed high level MSN method and radiomics textural features on the MDD and BD clinical studies.
Collapse
Affiliation(s)
- Kai Sun
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China; College of Medical Information and Artificial Intelligence & Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Guanmao Chen
- Medical Imaging Center, First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Chunchen Liu
- College of Medical Information and Artificial Intelligence & Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Zihan Chu
- College of Medical Information and Artificial Intelligence & Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Li Huang
- Medical Imaging Center, First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Zhou Li
- College of Medical Information and Artificial Intelligence & Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Shuming Zhong
- Department of Psychiatry, First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xiaoying Ye
- Department of Psychiatry, First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yingli Zhang
- Department of Psychiatry, First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yanbin Jia
- Department of Psychiatry, First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Jiyang Pan
- Department of Psychiatry, First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Guifei Zhou
- School of Information Science and Technology, Yunnan Normal University, Kunming, China.
| | - Zhenyu Liu
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China; CAS Key Laboratory of Molecular Imaging, Institute of Automation, Beijing, China; School of Artificial Intelligence, University of Chinese Academy of Science, Beijing, China.
| | - Changbin Yu
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China; College of Medical Information and Artificial Intelligence & Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| | - Ying Wang
- Medical Imaging Center, First Affiliated Hospital of Jinan University, Guangzhou, China.
| |
Collapse
|
43
|
Wang Y, Yu Y, Gu L, Sun Y, Yan J, Zhang H, Zhang Y. Radiomics feature is a risk factor for locally advanced cervical cancer treated using concurrent chemoradiotherapy based on magnetic resonance imaging: a retrospective study. BMC Cancer 2025; 25:230. [PMID: 39930343 PMCID: PMC11809009 DOI: 10.1186/s12885-025-13625-y] [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: 04/15/2024] [Accepted: 02/03/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND Although concurrent chemoradiotherapy (CCRT) is the standard treatment strategy for locally advanced cervical squamous carcinoma (LACSC), there are still individual differences. It is of vital importance to establish a radiomics-based model for predicting overall survival (OS) of LACSC patients treated using CCRT, and evaluating the feasibility of adjuvant chemotherapy (ACT). METHODS 122 LACSC patients were retrospectively analyzed who underwent pelvic MRI before standard CCRT between January 2013 and September 2016, including 85 patients in training set and 37 patients in testing set. 3D Slicer was used to segment images and extract features. IPMs software was used to select features and construct radscore. We selected the group with the largest area under the curves as the best result from 150 feature subsets and corresponding radscore. A nomogram was established using univariate and multivariate Cox analyses. We used Shapley Additive Explanations (SHAP) for further interpretation of the nomogram. Kaplan-Meier curves demonstrated the associations of radscore and clinical characteristics with OS and ACT. RESULTS Radscore was a prognostic factor (P = 0.001) which constructed using 10 radiomics features influencing the OS of patients with LACSC treated using CCRT. The radiomics-clinical model estimated OS (training, C-index: 0.761; testing, C-index: 0.718) more accurately than the clinical (training, C-index: 0.745; testing, C-index: 0.708) and radiomics models (training, C-index: 0.702; testing, C-index: 0.671). Radscore has the greatest impact on the prognosis of LACSC patients. We combined radscore and clinical factors to obtain risk scores. There was a better OS rate among low-risk patients than among high-risk patients (training, P = 0.034; testing, P = 0.003). Compared with CCRT, ACT + CCRT did not improve prognosis (high-risk patients, P = 0.703; all patients, P = 0.425). CONCLUSIONS Radscore independently predicted OS in LACSC. The radiomics-clinical nomogram improved individualized OS estimation. Patients did not benefit from ACT.
Collapse
Affiliation(s)
- Yuan Wang
- Department of Gynecological Radiotherapy, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150081, Heilongjiang Province, China
| | - Yanyan Yu
- Department of Radiology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Lina Gu
- Department of Gynecological Radiotherapy, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150081, Heilongjiang Province, China
| | - Yunfeng Sun
- Department of Radiology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Jiazhuo Yan
- Department of Gynecological Radiotherapy, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150081, Heilongjiang Province, China
| | - Hongxia Zhang
- Department of Radiology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yunyan Zhang
- Department of Gynecological Radiotherapy, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150081, Heilongjiang Province, China.
| |
Collapse
|
44
|
Liang L, Pang JS, Gao RZ, Que Q, Wu YQ, Peng JB, Bai XM, Qin Q, Tang QQ, Li LP, He Y, Yang H. Development and validation of a combined radiomic and clinical model based on contrast-enhanced ultrasound for preoperative prediction of CK19-positive hepatocellular carcinoma. Abdom Radiol (NY) 2025:10.1007/s00261-025-04799-x. [PMID: 39907719 DOI: 10.1007/s00261-025-04799-x] [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: 11/23/2024] [Revised: 12/31/2024] [Accepted: 01/03/2025] [Indexed: 02/06/2025]
Abstract
PURPOSE We aimed to develop and validate a combined model integrating radiomic features derived from Contrast-Enhanced Ultrasound (CEUS) images and clinical parameters for preoperative prediction of CK19-positive status in hepatocellular carcinoma (HCC). METHODS A total of 434 patients who underwent CEUS and surgical resection from January 2020 to December 2023 were included. Patients were randomly divided into a training cohort (n = 304) and a validation cohort (n = 130). Radiomic features were extracted from multiphase CEUS images, including two-dimensional ultrasound (US), arterial, portal venous, and delayed phases, and combined to derive a Radscore model. Subsequently, a Combined Model was constructed using the Radscore and clinical parameters. Model performance was assessed using calibration, discrimination, and clinical utility. RESULTS Multivariate logistic regression analysis identified Radscore (OR = 10.054, 95% CI: 5.931-19.120, p < 0.001) and AFP levels > 200 ng/mL (OR = 5.027, 95% CI: 2.089-12.784, p < 0.001) as significant predictors in the combined model. The AUC (Area Under the Curve) for the Combined Model was 0.954 in the training cohort and 0.927 in the validation cohort, compared to 0.939 and 0.917 for the Radscore Model alone. Calibration curves demonstrated strong concordance between predicted and actual outcomes. Decision curve analysis (DCA) showed that both the Radscore Model and the Combined Model exhibited good net benefits across a wide range of threshold values in both the training and validation cohorts. CONCLUSION The Radscore based on CEUS, combined with the serum markers AFP > 200 ng/L to construct a Combined Model, shows good predictive performance for CK19 + hepatocellular carcinoma (HCC).
Collapse
Affiliation(s)
- Li Liang
- Department of Medical Ultrasound, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Jin-Shu Pang
- Department of Medical Ultrasound, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Rui-Zhi Gao
- Department of Medical Ultrasound, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Qiao Que
- Department of Medical Ultrasound, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Yu-Quan Wu
- Department of Medical Ultrasound, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Jin-Bo Peng
- Department of Medical Ultrasound, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Xiu-Mei Bai
- Department of Medical Ultrasound, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Qiong Qin
- Department of Medical Ultrasound, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Quan-Quan Tang
- Department of Medical Ultrasound, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Li-Peng Li
- Department of Medical Ultrasound, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Yun He
- Department of Medical Ultrasound, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, Guangxi Zhuang Autonomous Region, China.
| | - Hong Yang
- Department of Medical Ultrasound, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, Guangxi Zhuang Autonomous Region, China.
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, China.
- Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor/Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Nanning, China.
| |
Collapse
|
45
|
Sghedoni R, Origgi D, Cucurachi N, Minischetti GC, Alio D, Savini G, Botta F, Marzi S, Aiello M, Rancati T, Cusumano D, Politi LS, Didonna V, Massafra R, Petrillo A, Esposito A, Imparato S, Anemoni L, Bortolotto C, Preda L, Boldrini L. Stability of radiomic features in magnetic resonance imaging of the female pelvis: A multicentre phantom study. Phys Med 2025; 130:104895. [PMID: 39793255 DOI: 10.1016/j.ejmp.2025.104895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 12/18/2024] [Accepted: 01/03/2025] [Indexed: 01/13/2025] Open
Affiliation(s)
- Roberto Sghedoni
- Medical Physics Unit, Azienda USL - IRCCS di Reggio Emilia, Viale Risorgimento 80, Reggio Emilia, Italy.
| | - Daniela Origgi
- Medical Physics Unit, IEO, European Institute of Oncology, IRCCS, Via Ripamonti 435, Milano, Italy
| | - Noemi Cucurachi
- Medical Physics Unit, Azienda USL - IRCCS di Reggio Emilia, Viale Risorgimento 80, Reggio Emilia, Italy
| | - Giuseppe Castiglioni Minischetti
- Medical Physics Unit, IEO, European Institute of Oncology, IRCCS, Via Ripamonti 435, Milano, Italy; School of Medical Physics, University of Milan, Via Celoria 16, 20133 Milan, Italy
| | - Davide Alio
- Medical Physics Unit, IEO, European Institute of Oncology, IRCCS, Via Ripamonti 435, Milano, Italy; School of Medical Physics, University of Milan, Via Celoria 16, 20133 Milan, Italy
| | - Giovanni Savini
- Department of Biomedical Sciences, Humanitas University, Via R. Levi Montalcini 4, 20072, Pieve Emanuele, Milan, Italy; Neuroradiology Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Francesca Botta
- Medical Physics Unit, IEO, European Institute of Oncology, IRCCS, Via Ripamonti 435, Milano, Italy
| | - Simona Marzi
- Medical Physics Laboratory, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144 Roma, Italy
| | - Marco Aiello
- IRCCS SYNLAB SDN, Via Francesco Crispi, 8, 80121 Napoli, Italy
| | - Tiziana Rancati
- Data Science Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Giacomo Venezian, 1, 20133 Milano, Italy
| | - Davide Cusumano
- UO Fisica Medica e Radioprotezione, Mater Olbia Hospital, SS 125 Orientale Sarda, 07026 Olbia, Italy
| | - Letterio Salvatore Politi
- Department of Biomedical Sciences, Humanitas University, Via R. Levi Montalcini 4, 20072, Pieve Emanuele, Milan, Italy; Neuroradiology Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Vittorio Didonna
- I.R.C.C.S. Istituto Tumori "Giovanni Paolo II", Viale Orazio Flacco 65, Bari 70124, Italy
| | - Raffaella Massafra
- I.R.C.C.S. Istituto Tumori "Giovanni Paolo II", Viale Orazio Flacco 65, Bari 70124, Italy
| | - Antonella Petrillo
- Istituto Nazionale Tumori IRCCS Fondazione Pascale, Via M. Semmola, 52, 80131 Napoli, Italy
| | - Antonio Esposito
- Experimetal Imaging Center, IRCCS San Raffaele Scientific Institute, Via Olgettina, 60, 20132 Milano, Italy; Vita-Salute San Raffaele University, School of Medicine, Via Olgettina, 58, 20132 Milano, Italy
| | - Sara Imparato
- Unità di Diagnostica per Immagini, CNAO, Via Erminio Borloni, 1, 27100 Pavia, Italy
| | - Luca Anemoni
- Unità di Diagnostica per Immagini, CNAO, Via Erminio Borloni, 1, 27100 Pavia, Italy
| | - Chandra Bortolotto
- Diagnostic Imaging and Radiotherapy Unit, Department of Clinical, Surgical, Diagnostic, and Pediatric Sciences, University of Pavia, Viale Golgi 19, 27100 Pavia, Italy; Radiology Institute, Fondazione IRCCS Policlinico San Matteo, Viale Golgi 19, 27100 Pavia, Italy
| | - Lorenzo Preda
- Diagnostic Imaging and Radiotherapy Unit, Department of Clinical, Surgical, Diagnostic, and Pediatric Sciences, University of Pavia, Viale Golgi 19, 27100 Pavia, Italy; Radiology Institute, Fondazione IRCCS Policlinico San Matteo, Viale Golgi 19, 27100 Pavia, Italy
| | - Luca Boldrini
- Dipartimento di Diagnostica per Immagini e Radioterapia Oncologica, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Largo Agostino Gemelli 8, 00168 Roma, Italy
| |
Collapse
|
46
|
Li H, Dong D, Fang M, He B, Liu S, Hu C, Liu Z, Wang H, Tang L, Tian J. ContraSurv: Enhancing Prognostic Assessment of Medical Images via Data-Efficient Weakly Supervised Contrastive Learning. IEEE J Biomed Health Inform 2025; 29:1232-1242. [PMID: 39437290 DOI: 10.1109/jbhi.2024.3484991] [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: 10/25/2024]
Abstract
Prognostic assessment remains a critical challenge in medical research, often limited by the lack of well-labeled data. In this work, we introduce ContraSurv, a weakly-supervised learning framework based on contrastive learning, designed to enhance prognostic predictions in 3D medical images. ContraSurv utilizes both the self-supervised information inherent in unlabeled data and the weakly-supervised cues present in censored data, refining its capacity to extract prognostic representations. For this purpose, we establish a Vision Transformer architecture optimized for our medical image datasets and introduce novel methodologies for both self-supervised and supervised contrastive learning for prognostic assessment. Additionally, we propose a specialized supervised contrastive loss function and introduce SurvMix, a novel data augmentation technique for survival analysis. Evaluations were conducted across three cancer types and two imaging modalities on three real-world datasets. The results confirmed the enhanced performance of ContraSurv over competing methods, particularly in data with a high censoring rate.
Collapse
|
47
|
Yan T, Yan Z, Chen G, Xu S, Wu C, Zhou Q, Wang G, Li Y, Jia M, Zhuang X, Yang J, Liu L, Wang L, Wu Q, Wang B, Yan T. Survival outcome prediction of esophageal squamous cell carcinoma patients based on radiomics and mutation signature. Cancer Imaging 2025; 25:9. [PMID: 39891186 PMCID: PMC11783911 DOI: 10.1186/s40644-024-00821-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 12/29/2024] [Indexed: 02/03/2025] Open
Abstract
BACKGROUND The present study aimed to develop a nomogram model for predicting overall survival (OS) in esophageal squamous cell carcinoma (ESCC) patients. METHODS A total of 205 patients with ESCC were enrolled and randomly divided into a training cohort (n = 153) and a test cohort (n = 52) at a ratio of 7:3. Multivariate Cox regression was used to construct the radiomics model based on CT data. The mutation signature was constructed based on whole genome sequencing data and found to be significantly associated with the prognosis of patients with ESCC. A nomogram model combining the Rad-score and mutation signature was constructed. An integrated nomogram model combining the Rad-score, mutation signature, and clinical factors was constructed. RESULTS A total of 8 CT features were selected for multivariate Cox regression analysis to determine whether the Rad-score was significantly correlated with OS. The area under the curve (AUC) of the radiomics model was 0.834 (95% CI, 0.767-0.900) for the training cohort and 0.733 (95% CI, 0.574-0.892) for the test cohort. The Rad-score, S3, and S6 were used to construct an integrated RM nomogram. The predictive performance of the RM nomogram model was better than that of the radiomics model, with an AUC of 0. 830 (95% CI, 0.761-0.899) in the training cohort and 0.793 (95% CI, 0.653-0.934) in the test cohort. The Rad-score, TNM stage, lymph node metastasis status, S3, and S6 were used to construct an integrated RMC nomogram. The predictive performance of the RMC nomogram model was better than that of the radiomics model and RM nomogram model, with an AUC of 0. 862 (95% CI, 0.795-0.928) in the training cohort and 0. 837 (95% CI, 0.705-0.969) in the test cohort. CONCLUSION An integrated nomogram model combining the Rad-score, mutation signature, and clinical factors can better predict the prognosis of patients with ESCC.
Collapse
Affiliation(s)
- Ting Yan
- Second Clinical Medical College, Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Zhenpeng Yan
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Guohui Chen
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Songrui Xu
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Chenxuan Wu
- School of Life Science, Beijing Institute of Technology, Beijing, People's Republic of China
| | - Qichao Zhou
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Guolan Wang
- School of Computer Information Engineering, Shanxi Technology and Business University, Taiyuan, Shanxi, 030006, People's Republic of China
| | - Ying Li
- College of Information and Computer, Taiyuan University of Technology, Taiyuan, Shanxi, 030024, People's Republic of China
| | - Mengjiu Jia
- School of Computer Information Engineering, Shanxi Technology and Business University, Taiyuan, Shanxi, 030006, People's Republic of China
| | - Xiaofei Zhuang
- Department of Thoracic Surgery, Shanxi Cancer Hospital, Taiyuan, Shanxi, 030013, People's Republic of China
| | - Jie Yang
- Department of Gastroenterology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Lili Liu
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Lu Wang
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Qinglu Wu
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Bin Wang
- College of Information and Computer, Taiyuan University of Technology, Taiyuan, Shanxi, 030024, People's Republic of China.
| | - Tianyi Yan
- School of Life Science, Beijing Institute of Technology, Beijing, People's Republic of China.
| |
Collapse
|
48
|
Huang Q, Huang F, Chen C, Xiao P, Liu J, Gao Y. Machine-learning model based on ultrasomics for non-invasive evaluation of fibrosis in IgA nephropathy. Eur Radiol 2025:10.1007/s00330-025-11368-9. [PMID: 39853332 DOI: 10.1007/s00330-025-11368-9] [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: 06/05/2024] [Revised: 12/02/2024] [Accepted: 12/19/2024] [Indexed: 01/26/2025]
Abstract
OBJECTIVES To develop and validate an ultrasomics-based machine-learning (ML) model for non-invasive assessment of interstitial fibrosis and tubular atrophy (IF/TA) in patients with IgA nephropathy (IgAN). MATERIALS AND METHODS In this multi-center retrospective study, 471 patients with primary IgA nephropathy from four institutions were included (training, n = 275; internal testing, n = 69; external testing, n = 127; respectively). The least absolute shrinkage and selection operator logistic regression with tenfold cross-validation was used to identify the most relevant features. The ML models were constructed based on ultrasomics. The Shapley Additive Explanation (SHAP) was used to explore the interpretability of the models. Logistic regression analysis was employed to combine ultrasomics, clinical data, and ultrasound imaging characteristics, creating a comprehensive model. A receiver operating characteristic curve, calibration, decision curve, and clinical impact curve were used to evaluate prediction performance. RESULTS To differentiate between mild and moderate-to-severe IF/TA, three prediction models were developed: the Rad_SVM_Model, Clinic_LR_Model, and Rad_Clinic_Model. The area under curves of these three models were 0.861, 0.884, and 0.913 in the training cohort, and 0.760, 0.860, and 0.894 in the internal validation cohort, as well as 0.794, 0.865, and 0.904 in the external validation cohort. SHAP identified the contribution of radiomics features. Difference analysis showed that there were significant differences between radiomics features and fibrosis. The comprehensive model was superior to that of individual indicators and performed well. CONCLUSIONS We developed and validated a model that combined ultrasomics, clinical data, and clinical ultrasonic characteristics based on ML to assess the extent of fibrosis in IgAN. KEY POINTS Question Currently, there is a lack of a comprehensive ultrasomics-based machine-learning model for non-invasive assessment of the extent of Immunoglobulin A nephropathy (IgAN) fibrosis. Findings We have developed and validated a robust and interpretable machine-learning model based on ultrasomics for assessing the degree of fibrosis in IgAN. Clinical relevance The machine-learning model developed in this study has significant interpretable clinical relevance. The ultrasomics-based comprehensive model had the potential for non-invasive assessment of fibrosis in IgAN, which helped evaluate disease progress.
Collapse
Affiliation(s)
- Qun Huang
- Department of Ultrasound, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Fangyi Huang
- Department of Ultrasound, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Chengcai Chen
- Department of Ultrasound, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Pan Xiao
- Department of Ultrasound, Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jiali Liu
- Department of Ultrasound, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Yong Gao
- Department of Ultrasound, First Affiliated Hospital of Guangxi Medical University, Nanning, China.
| |
Collapse
|
49
|
Wang Z, Qiu J, Shen X, Yang F, Liu X, Wang X, Ke N. A nomogram to preoperatively predict the aggressiveness of pancreatic neuroendocrine tumors based on CT features and 3D CT radiomic features. Abdom Radiol (NY) 2025:10.1007/s00261-024-04759-x. [PMID: 39841226 DOI: 10.1007/s00261-024-04759-x] [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: 12/09/2024] [Accepted: 12/10/2024] [Indexed: 01/23/2025]
Abstract
OBJECTIVES Combining Computed Tomography (CT) intuitive anatomical features with Three-Dimensional (3D) CT multimodal radiomic imaging features to construct a model for assessing the aggressiveness of pancreatic neuroendocrine tumors (pNETs) prior to surgery. METHODS This study involved 242 patients, randomly assigned to training (170) and validation (72) cohorts. Preoperative CT and 3D CT radiomic features were used to develop a model predicting pNETs aggressiveness. The aggressiveness of pNETs was characterized by a combination of factors including G3 grade, nodal involvement (N + status), presence of distant metastases, and/or recurrence of the disease. RESULTS Three distinct predictive models were constructed to evaluate the aggressiveness of pNETs using CT features, 3D CT radiomic features, and their combination. The combined model demonstrated the greatest predictive accuracy and clinical applicability in both the training and validation sets (AUCs (95% CIs) = 0.93 (0.90-0.97) and 0.89 (0.79-0.98), respectively). Subsequently, a nomogram was developed using the features from the combined model, displaying strong alignment between actual observations and predictions as indicated by the calibration curves. Using a nomogram score of 86.06, patients were classified into high- and low-aggressiveness groups, with the high-aggressiveness group demonstrating poorer overall survival and shorter disease-free survival. CONCLUSION This study presents a combined model incorporating CT and 3D CT radiomic features, which accurately predicts the aggressiveness of PNETs preoperatively.
Collapse
Affiliation(s)
- Ziyao Wang
- West China Hospital of Sichuan University, Chengdu, China
| | - Jiajun Qiu
- West China Hospital of Sichuan University, Chengdu, China
| | - Xiaoding Shen
- West China Hospital of Sichuan University, Chengdu, China
| | - Fan Yang
- West China Hospital of Sichuan University, Chengdu, China
| | - Xubao Liu
- West China Hospital of Sichuan University, Chengdu, China
| | - Xing Wang
- West China Hospital of Sichuan University, Chengdu, China.
| | - Nengwen Ke
- West China Hospital of Sichuan University, Chengdu, China.
| |
Collapse
|
50
|
Yan M, Zhang Z, Tian J, Yu J, Dekker A, Ruysscher DD, Wee L, Zhao L. Whole lung radiomic features are associated with overall survival in patients with locally advanced non-small cell lung cancer treated with definitive radiotherapy. Radiat Oncol 2025; 20:9. [PMID: 39825409 PMCID: PMC11742218 DOI: 10.1186/s13014-025-02583-1] [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: 04/30/2024] [Accepted: 01/03/2025] [Indexed: 01/20/2025] Open
Abstract
BACKGROUND Several studies have suggested that lung tissue heterogeneity is associated with overall survival (OS) in lung cancer. However, the quantitative relationship between the two remains unknown. The purpose of this study is to investigate the prognostic value of whole lung-based and tumor-based radiomics for OS in LA-NSCLC treated with definitive radiotherapy. METHODS A total of 661 patients with LA-NSCLC treated with definitive radiotherapy in combination with chemotherapy were enrolled in this study, with 292 patients in the training set, 57 patients from the same hospital from January to December 2017 as an independent test set (test-set-1), 83 patients from a multi-institutional prospective clinical trial data set (RTOG0617) as test-set-2, and 229 patients from a Dutch radiotherapy center as test-set-3. Tumor-based radiomic features and whole lung-based radiomic features were extracted from primary tumor and whole lungs (excluding the primary tumor) delineations in planning CT images. Feature selection of radiomic features was done by the least absolute shrinkage (LASSO) method embedded with a Cox proportional hazards (CPH) model with 5-fold cross-internal validation, with 1000 bootstrap samples. Radiomics prognostic scores (RS) were calculated by CPH regression based on selected features. Three models based on a tumor RS, and a lung RS separately and their combinations were constructed. The Harrell concordance index (C-index) and calibration curves were used to evaluate the discrimination and calibration performance. Patients were stratified into high and low risk groups based on median RS, and a log-rank test was performed. RESULTS The discrimination ability of lung- and tumor-based radiomics model was similar in terms of C-index, 0.69 vs. 0.68 in training set, 0.68 vs. 0.66 in test-set-1, 0.61 vs. 0.62 in test-set-2, 0.65 vs. 0.64 in test-set-3. The combination of tumor- and lung-based radiomics model performed best, with C-index of 0.71 in training set, 0.70 in test-set-1, 0.69 in test-set-2, and 0.68 in test-set-3. The calibration curve showed good agreement between predicted values and actual values. Patients were well stratified in training set, test-set-1 and test-set-3. In test-set-2, it was only whole lung-based RS that could stratify patients well and tumor-based RS performed bad. CONCLUSION Lung- and tumor-based radiomic features have the power to predict OS in LA-NSCLC. The combination of tumor- and lung-based radiomic features can achieve optimal performance.
Collapse
Affiliation(s)
- Meng Yan
- Department of Radiation Oncology, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Zhen Zhang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China.
- Department of Radiation Oncology (Maastro), GROW Research Institute for Oncology and Reproduction, Maastricht University Medical Centre+, Maastricht, The Netherlands.
| | - Jia Tian
- Department of Radiation Oncology, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Jiaqi Yu
- 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, 100021, China
| | - Andre Dekker
- Department of Radiation Oncology (Maastro), GROW Research Institute for Oncology and Reproduction, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Dirk de Ruysscher
- Department of Radiation Oncology (Maastro), GROW Research Institute for Oncology and Reproduction, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Leonard Wee
- Department of Radiation Oncology (Maastro), GROW Research Institute for Oncology and Reproduction, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Lujun Zhao
- Department of Radiation Oncology, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.
| |
Collapse
|