1
|
Pisani N, Destito M, Ricciardi C, Pellecchia MT, Cesarelli M, Esposito F, Spadea MF, Amato F. Repeatability of radiomic features from brain T1-W MRI after image intensity normalization: Implications for longitudinal studies on structural neurodegeneration. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2025; 265:108738. [PMID: 40203781 DOI: 10.1016/j.cmpb.2025.108738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 12/27/2024] [Accepted: 03/22/2025] [Indexed: 04/11/2025]
Abstract
BACKGROUND AND OBJECTIVE Radiomics extracts quantitative features from magnetic resonance images (MRI) and is especially useful in the presence of subtle pathological changes within human soft tissues. This scenario, however, may not cover the effects of intrinsic, e.g., aging-related, (physiological) neurodegeneration of normal brain tissue. The aim of the work was to study the repeatability of radiomic features extracted from an apparently normal area in longitudinally acquired T1-weighted MR brain images using three different intensity normalization approaches typically used in radiomics: Z-score, WhiteStripe and Nyul. METHODS Fifty-nine images of hearing impaired, yet cognitively intact, patients were repeatedly acquired in two different time points within six months. Ninety-one radiomic features were obtained from an area within the pons region, considered to be a healthy brain tissue according to previous analyses and reports. The Intraclass Correlation Coefficient (ICC) and the Concordance Correlation Coefficient (CCC) in the repeatability study were used as metrics. RESULTS Features extracted from the MRI normalized with Z-score showed results comparable in both ICC (0.90 (0.82-0.98)) and CCC (0.82 (0.69-0.95)) distribution values, in terms of median and quartiles, with those extracted from the images normalized with WhiteStripe (0.89 (0.84-0.92)) and (0.80 (0.73-0.84)), respectively. CONCLUSION Our findings underline the importance of, providing useful guidelines for, the intensity normalization of brain MRI prior to a longitudinal radiomic analysis.
Collapse
Affiliation(s)
- Noemi Pisani
- Department of Advanced Biomedical Sciences, University of Naples Federico II, 80131 Naples, Italy.
| | - Michela Destito
- Department of Experimental and Clinical Medicine, University of Catanzaro, 88100 Catanzaro, Italy.
| | - Carlo Ricciardi
- Department of Electrical Engineering and Information Technology, University of Naples Federico II, 80125 7 Naples, Italy.
| | - Maria Teresa Pellecchia
- Department of Medicine, Surgery and Dentistry 12 "Scuola Medica Salernitana", University of Salerno, 84131 Salerno, Italy.
| | - Mario Cesarelli
- Department of Engineering, University of Sannio, 82100 Benevento, Italy.
| | - Fabrizio Esposito
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 10 Naples, Italy.
| | - Maria Francesca Spadea
- Institute of Biomedical Engineering, Karlsruhe Institute of Technology (KIT), 76131 Karlsruhe, Germany.
| | - Francesco Amato
- Department of Electrical Engineering and Information Technology, University of Naples Federico II, 80125 7 Naples, Italy.
| |
Collapse
|
2
|
Schwarzhans F, George G, Escudero Sanchez L, Zaric O, Abraham JE, Woitek R, Hatamikia S. Image normalization techniques and their effect on the robustness and predictive power of breast MRI radiomics. Eur J Radiol 2025; 187:112086. [PMID: 40184762 DOI: 10.1016/j.ejrad.2025.112086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 03/20/2025] [Accepted: 03/28/2025] [Indexed: 04/07/2025]
Abstract
BACKGROUND AND PURPOSE Radiomics analysis has emerged as a promising approach to aid in cancer diagnosis and treatment. However, radiomics research currently lacks standardization, and radiomics features can be highly dependent on acquisition and pre-processing techniques used. In this study, we aim to investigate the effect of various image normalization techniques on robustness of radiomics features extracted from breast cancer patient MRI scans. MATERIALS AND METHODS MRI scans from the publicly available MAMA-MIA dataset and an internal breast MRI test set depicting triple negative breast cancer (TNBC) were used. We compared the effect of commonly used image normalization techniques on radiomics feature robustnessusing Concordance-Correlation-Coefficient (CCC) between multiple combinations of normalization approaches. We also trained machine learning-based prediction models of pathologic complete response (pCR) on radiomics after different normalization techniques were used and compared their areas under the receiver operating characteristic curve (ROC-AUC). RESULTS For predicting complete pathological response from pre-treatment breast cancer MRI radiomics, the highest overall ROC-AUC was achieved by using a combination of three different normalization techniques indicating their potentially powerful role when working with heterogeneous imaging data. The effect of normalization was more pronounced with smaller training data and normalization may be less important with increasing abundance of training data. Additionally, we observed considerable differences between MRI data sets and their feature robustness towards normalization. CONCLUSION Overall, we were able to demonstrate the importance of selecting and standardizing normalization methods for accurate and reliable radiomics analysis in breast MRI scans especially with small training data sets.
Collapse
Affiliation(s)
- Florian Schwarzhans
- Research Center for Medical Image Analysis and Artificial Intelligence (MIAAI), Department of Medicine, Danube Private University, Krems, Rathausplatz 1, AT-3500 Krems-Stein, Austria.
| | - Geevarghese George
- Research Center for Medical Image Analysis and Artificial Intelligence (MIAAI), Department of Medicine, Danube Private University, Krems, Rathausplatz 1, AT-3500 Krems-Stein, Austria.
| | - Lorena Escudero Sanchez
- Department of Radiology, University of Cambridge, UK; Cancer Research UK Cambridge Centre, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK.
| | - Olgica Zaric
- Research Center for Medical Image Analysis and Artificial Intelligence (MIAAI), Department of Medicine, Danube Private University, Krems, Rathausplatz 1, AT-3500 Krems-Stein, Austria.
| | - Jean E Abraham
- Cancer Research UK Cambridge Centre, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK; Precision Breast Cancer Institute, Department of Oncology, University of Cambridge, Cambridge CB2 0QQ, UK.
| | - Ramona Woitek
- Research Center for Medical Image Analysis and Artificial Intelligence (MIAAI), Department of Medicine, Danube Private University, Krems, Rathausplatz 1, AT-3500 Krems-Stein, Austria; Department of Radiology, University of Cambridge, UK.
| | - Sepideh Hatamikia
- Research Center for Medical Image Analysis and Artificial Intelligence (MIAAI), Department of Medicine, Danube Private University, Krems, Rathausplatz 1, AT-3500 Krems-Stein, Austria; Austrian Center for Medical Innovation and Technology (ACMIT), Viktor Kaplan-Straße 2/1, 2700 Wiener Neustadt, Austria.
| |
Collapse
|
3
|
Ho WLJ, Fetisov N, Hall LO, Goldgof D, Schabath MB. Utilizing Clinicopathological and Radiomic Features for Risk Stratification of Lung Cancer Recurrence. Acad Radiol 2025:S1076-6332(25)00415-5. [PMID: 40379589 DOI: 10.1016/j.acra.2025.04.062] [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: 02/09/2025] [Revised: 04/21/2025] [Accepted: 04/24/2025] [Indexed: 05/19/2025]
Abstract
RATIONALE AND OBJECTIVES To predict recurrence risk in patients with surgically resected non-small cell lung cancer (NSCLC) using radiomic analysis and clinicopathological factors. MATERIALS AND METHODS 293 patients with surgically resected stage IA-IIIA NSCLC were analyzed. Patients were randomly stratified into development and test cohorts. The development cohort was further divided into training and validation subsets for feature selection and model building, then applied to the test cohort. Pre-treatment computed tomography were segmented and 107 pyRadiomics features were extracted from intratumoral and peritumoral regions. Feature selection was performed using the maximum relevance minimum redundancy algorithm and Lasso regression. Clinical covariates were selected using univariable Cox regression. Radiomic, clinical, and radiomic-clinical models were constructed using a logistic regression classifier and evaluated using area under the curve (AUC). Kaplan-Meier curves for 3-year recurrence-free survival were compared between high-risk and low-risk groups using the log-rank test. RESULTS 20 percent of patients experienced recurrence within 3 years. The radiomic-clinical model (AUC 0.77) outperformed the radiomic, clinical, and TNM stage models (AUC 0.76, 0.71, and 0.70, respectively) on the test set. Recurrence risk was five times higher in the high-risk group than the low-risk group (p<0.01) after stratification with the radiomic-clinical model. The most important features were regional lymph node metastases, the "GLDM Large Dependence Emphasis" texture, and the "Elongation" shape feature. CONCLUSION Radiomics analysis can be used in combination with clinicopathological features for effective recurrence risk stratification in patients with surgically resected NSCLC.
Collapse
Affiliation(s)
- Wai Lone J Ho
- University of South Florida, Morsani College of Medicine, Tampa, Florida (W.L.J.H.)
| | - Nikolai Fetisov
- Department of Computer Science and Engineering, University of South Florida, Tampa, Florida (N.F., L.O.H., D.G.)
| | - Lawrence O Hall
- Department of Computer Science and Engineering, University of South Florida, Tampa, Florida (N.F., L.O.H., D.G.)
| | - Dmitry Goldgof
- Department of Computer Science and Engineering, University of South Florida, Tampa, Florida (N.F., L.O.H., D.G.)
| | - Matthew B Schabath
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida (M.B.S.).
| |
Collapse
|
4
|
Xu J, Gao S, Zhu Q, Dai F, Sun C, Lee W, Ye Y, Deng G, Huang Z, Li X, Li J, Cheong S, Huang Q, Di J. Machine learning-based multiparametric CT radiomics for predicting microvascular invasion before nephrectomy in clear cell renal cell carcinoma. Abdom Radiol (NY) 2025:10.1007/s00261-025-04956-2. [PMID: 40249552 DOI: 10.1007/s00261-025-04956-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 04/10/2025] [Accepted: 04/11/2025] [Indexed: 04/19/2025]
Abstract
PURPOSE This study aimed to investigate the value of integrating computed tomography (CT)-based tumor radiomics features with clinical parameters for preoperative prediction of microvascular invasion (MVI) in clear cell renal cell carcinoma (ccRCC). METHODS We retrospectively analyzed data from a single-center cohort of ccRCC patients. Radiomics features were extracted from preoperative multiphasic CT scans (unenhanced, corticomedullary, and nephrographic phases). Following dimensionality reduction and feature selection, eight machine learning algorithms were evaluated to identify the optimal radiomics model. Independent clinical predictors were determined through univariate and multivariate analyses. A nomogram integrating the radiomics signature (rad-score) with significant clinical parameters was subsequently developed. Model performance was assessed using the area under the curve (AUC), decision curve analysis (DCA), and calibration curve analysis (CAC). RESULTS Of 143 initially enrolled patients, 110 met inclusion criteria after screening, with 5502 radiomics features extracted. The support vector classifier (SVM) model demonstrated the highest discriminative ability, achieving mean AUCs of 0.976 (training cohort) and 0.892 (test cohort), significantly outperforming the clinical model (training AUC = 0.935, test AUC = 0.933). The nomogram showed superior diagnostic performance, with AUCs of 0.958 (test). DCA and CAC confirmed its clinical utility and robustness. CONCLUSIONS Multiparametric CT radiomics models enable non-invasive prediction of MVI status in ccRCC, with the SVM-based algorithm showing optimal performance. The integrated nomogram provides excellent and consistent diagnostic accuracy, offering a valuable preoperative tool for clinical decision-making.
Collapse
Affiliation(s)
- Jinbin Xu
- Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Shuntian Gao
- Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Qin Zhu
- First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Fuyang Dai
- Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ciming Sun
- Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Weijen Lee
- Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yuedian Ye
- Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Gengguo Deng
- Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhansen Huang
- Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaoming Li
- Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jiang Li
- Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Samun Cheong
- Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Qunxiong Huang
- Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| | - Jinming Di
- Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
5
|
Lu J, Liu X, Ji X, Jiang Y, Zuo A, Guo Z, Yang S, Peng H, Sun F, Lu D. Predicting PD-L1 status in NSCLC patients using deep learning radiomics based on CT images. Sci Rep 2025; 15:12495. [PMID: 40216830 PMCID: PMC11992188 DOI: 10.1038/s41598-025-91575-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 02/21/2025] [Indexed: 04/14/2025] Open
Abstract
Radiomics refers to the utilization of automated or semi-automated techniques to extract and analyze numerous quantitative features from medical images, such as computerized tomography (CT) or magnetic resonance imaging (MRI) scans. This study aims to develop a deep learning radiomics (DLR)-based approach for predicting programmed death-ligand 1 (PD-L1) expression in patients with non-small cell lung cancer (NSCLC). Data from 352 NSCLC patients with known PD-L1 expression were collected, of which 48.29% (170/352) were tested positive for PD-L1 expression. Tumor regions of interest (ROI) were semi-automatically segmented based on CT images, and DL features were extracted using Residual Network 50. The least absolute shrinkage and selection operator (LASSO) algorithm was used for feature selection and dimensionality reduction. Seven algorithms were used to build models, and the most optimal ones were identified. A combined model integrating DLR with clinical data was also developed. The predictive performance of each model was evaluated using the area under the curve (AUC) of the receiver operating characteristic (ROC) curve analysis. The DLR model, based on CT images, demonstrated an AUC of 0.85 (95% confidence interval (CI), 0.82-0.88), sensitivity of 0.80 (0.74-0.85), and specificity of 0.73 (0.70-0.77) for predicting PD-L1 status. The integrated model exhibited superior performance, with an AUC of 0.91 (0.87-0.95), sensitivity of 0.85 (0.82-0.89), and specificity of 0.75 (0.72-0.80). Our findings indicate that the DLR model holds promise as a valuable tool for predicting the PD-L1 status in patients with NSCLC, which can greatly assist in clinical decision-making and the selection of personalized treatment strategies.
Collapse
Affiliation(s)
- Jiameng Lu
- Department of Respiratory, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Shandong Institute of Anesthesia and Respiratory Critical Medicine, 16766 Jingshilu, Lixia, Jinan, 250014, Shandong, People's Republic of China
- Faculty of Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, 999078, Macau Special Administrative Region, People's Republic of China
| | - Xinyi Liu
- Department of Respiratory, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Shandong Institute of Anesthesia and Respiratory Critical Medicine, 16766 Jingshilu, Lixia, Jinan, 250014, Shandong, People's Republic of China
| | - Xiaoqing Ji
- Department of Nursing, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, Shandong, China
| | - Yunxiu Jiang
- Department of Respiratory, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Shandong Institute of Anesthesia and Respiratory Critical Medicine, 16766 Jingshilu, Lixia, Jinan, 250014, Shandong, People's Republic of China
| | - Anli Zuo
- Department of Respiratory, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Shandong Institute of Anesthesia and Respiratory Critical Medicine, 16766 Jingshilu, Lixia, Jinan, 250014, Shandong, People's Republic of China
| | - Zihan Guo
- Department of Respiratory, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Shandong Institute of Anesthesia and Respiratory Critical Medicine, 16766 Jingshilu, Lixia, Jinan, 250014, Shandong, People's Republic of China
| | - Shuran Yang
- Department of Respiratory, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Shandong Institute of Anesthesia and Respiratory Critical Medicine, 16766 Jingshilu, Lixia, Jinan, 250014, Shandong, People's Republic of China
| | - Haiying Peng
- Department of Respiratory and Critical Care Medicine, The Second People's Hospital of Yibin City, 644002, Yibin, People's Republic of China
| | - Fei Sun
- Department of Respiratory and Critical Care Medicine, Jining No.1 People's Hospital, 272000, Jining, People's Republic of China
| | - Degan Lu
- Department of Respiratory, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Shandong Institute of Anesthesia and Respiratory Critical Medicine, 16766 Jingshilu, Lixia, Jinan, 250014, Shandong, People's Republic of China.
| |
Collapse
|
6
|
Kinoshita S, Nakaura T, Yoshizumi T, Itoh S, Ide T, Noshiro H, Hamada T, Kuroki T, Takami Y, Nagano H, Nanashima A, Endo Y, Utsunomiya T, Kajiwara M, Miyoshi A, Sakoda M, Okamoto K, Beppu T, Takatsuki M, Noritomi T, Baba H, Eguchi S. Real-world efficacy of radiomics versus clinical predictors for microvascular invasion in patients with hepatocellular carcinoma: Large cohort study. Hepatol Res 2025; 55:567-576. [PMID: 40317657 DOI: 10.1111/hepr.14149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 10/29/2024] [Accepted: 11/24/2024] [Indexed: 05/07/2025]
Abstract
AIM Microvascular invasion (MVI) affects the prognosis and treatment of hepatocellular carcinoma (HCC); however, its preoperative diagnosis is challenging. Analysis of computed tomography (CT) images using radiomics can detect MVI, but its effectiveness depends on the imaging conditions. We compared the efficacies of radiomics, clinical, and combined models for predicting MVI in HCC using nonstandardized scanning protocols. METHODS This multicenter study included 533 patients who underwent hepatic resection for HCC. Patients were divided randomly into training (n = 426) and test groups (n = 107). We manually extracted 3D CT features in hepatic arterial, portal venous, and venous phases. The radiomics model was trained by machine learning. A logistic regression model was developed based on clinical information, and a fused model was created integrating clinical information and radiomics prediction score (Rad_Score). We calculated areas under the receiver operating characteristic curves (AUCs) for the radiomics, clinical, and mixed models in the test groups. RESULTS The clinical model incorporated hepatitis B virus surface antigen, tumor diameter, and log-transformed α-fetoprotein and des-gamma-carboxyprothrombin. The AUCs of the radiomics and clinical models were comparable (p = 0.76). Rad_Score was not an independent significant factor in the fused model (p = 0.40) and its addition did not improve the accuracy of the clinical model alone (p = 0.51). CONCLUSIONS A clinical model is as effective as a CT radiomics model for predicting MVI status in patients with HCC based on real-world scanning data, and integration of both models does not improve the predictive performance compared with a clinical model alone.
Collapse
Affiliation(s)
- Shotaro Kinoshita
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Takeshi Nakaura
- Department of Diagnostic Radiology, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Tomoharu Yoshizumi
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shinji Itoh
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takao Ide
- Department of Surgery, Saga University Faculty of Medicine, Saga, Japan
| | - Hirokazu Noshiro
- Department of Surgery, Saga University Faculty of Medicine, Saga, Japan
| | - Takashi Hamada
- Department of Surgery, NHO Nagasaki Medical Center, Nagasaki, Japan
| | - Tamotsu Kuroki
- Department of Surgery, NHO Nagasaki Medical Center, Nagasaki, Japan
| | - Yuko Takami
- Department of Hepato-Biliary-Pancreatic Surgery, Clinical Research Institute, NHO Kyushu Medical Center, Fukuoka, Japan
| | - Hiroaki Nagano
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Atsushi Nanashima
- Division of Hepato-Biliary-Pancreas Surgery, Department of Surgery, University of Miyazaki Faculty of Medicine, Miyazaki, Japan
| | - Yuichi Endo
- Department of Gastroenterological and Pediatric Surgery, Oita University Faculty of Medicine, Oita, Japan
| | - Tohru Utsunomiya
- Department of Gastroenterological Surgery, Oita Prefectural Hospital, Oita, Japan
| | - Masatoshi Kajiwara
- Department of Gastroenterological Surgery, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Atsushi Miyoshi
- Department of Surgery, Saga-Ken Medical Centre Koseikan, Saga, Japan
| | - Masahiko Sakoda
- Department of Surgery, Kagoshima Kouseiren Hospital, Kagoshima, Japan
| | - Kohji Okamoto
- Department of Surgery, Gastroenterology and Hepatology Center, Kitakyushu City Yahata Hospital, Kitakyushu, Japan
| | - Toru Beppu
- Department of Surgery, Yamaga City Medical Center, Yamaga, Japan
| | - Mitsuhisa Takatsuki
- Department of Digestive and General Surgery, Graduate School of Medicine, University of the Ryukyus, Nishihara, Japan
| | - Tomoaki Noritomi
- Department of Surgery, Fukuoka Tokushukai Hospital, Fukuoka, Japan
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Susumu Eguchi
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
7
|
Deng Y, Yu H, Duan X, Liu L, Huang Z, Song B. A CT-based radiomics nomogram for the preoperative prediction of perineural invasion in pancreatic ductal adenocarcinoma. Front Oncol 2025; 15:1525835. [PMID: 40104508 PMCID: PMC11913684 DOI: 10.3389/fonc.2025.1525835] [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: 11/10/2024] [Accepted: 02/06/2025] [Indexed: 03/20/2025] Open
Abstract
Purpose To develop a nomogram based on CT radiomics features for preoperative prediction of perineural invasion (PNI) in pancreatic ductal adenocarcinoma (PDAC) patients. Methods A total of 217 patients with histologically confirmed PDAC were enrolled in this retrospective study. Radiomics features were extracted from the whole tumor. Univariate analysis, least absolute shrinkage and selection operator and logistic regression were applied for feature selection and radiomics model construction. Finally, a nomogram combining the radiomics score (Rad-score) and clinical characteristics was established. Receiver operating characteristic curve analysis, calibration curve analysis and decision curve analysis (DCA) were used to evaluate the predictive performance of the nomogram. Results According to multivariate analysis, CT features, including the radiologists evaluated PNI status based on CECT (CTPNI) (OR=1.971 [95% CI: 1.165, 3.332], P=0.01), the lymph node status determined on CECT (CTLN) (OR=2.506 [95%: 1.416, 4.333], P=0.001) and the Rad-score (OR=3.666 [95% CI: 2.069, 6.494], P<0.001), were significantly associated with PNI. The area under the receiver operating characteristic curve (AUC) for the nomogram combined with the Rad-score, CTLN and CTPNI achieved favorable discrimination of PNI status, with AUCs of 0.846 and 0.778 in the training and testing cohorts, respectively, which were superior to those of the Rad-score (AUC of 0.720 in the training cohort and 0.640 in the testing cohort) and CTPNI (AUC of 0.610 in the training cohort and 0.675 in the testing cohort). The calibration plot and decision curve showed good results. Conclusion The CT-based radiomics nomogram has the potential to accurately predict PNI in patients with PDAC.
Collapse
Affiliation(s)
- Yan Deng
- Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
- Department of Radiology, Sichuan Key Laboratory of Medical Imaging, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Haopeng Yu
- Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
| | - Xiuping Duan
- Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
| | - Li Liu
- Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
| | - Zixing Huang
- Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
| | - Bin Song
- Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
- Department of Radiology, Sanya People's Hospital, Sanya, Hainan, China
| |
Collapse
|
8
|
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
|
9
|
Xu G, Feng F, Cui Y, Fu Y, Xiao Y, Chen W, Li M. Prediction of postoperative disease-free survival in colorectal cancer patients using CT radiomics nomogram: a multicenter study. Acta Radiol 2025; 66:269-280. [PMID: 39894908 DOI: 10.1177/02841851241302521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
BackgroundRadiomics analysis is widely used to assess tumor prognosis.PurposeTo explore the value of computed tomography (CT) radiomics nomogram in predicting disease-free survival (DFS) of patients with colorectal cancer (CRC) after operation.Material and MethodsA total of 522 CRC patients from three centers were retrospectively included. Radiomics features were extracted from CT images, and the least absolute shrinkage and selection operator Cox regression algorithm was employed to select radiomics features. Clinical risk factors associated with DFS were selected through univariate and multivariate Cox regression analysis to build the clinical model. A predictive nomogram was developed by amalgamating pertinent clinical risk factors and radiomics features. The predictive performance of the nomogram was evaluated using the C-index, calibration curve, and decision curve. DFS probabilities were estimated using the Kaplan-Meier method.ResultsIntegrating the retained eight radiomics features and three clinical risk factors (pathological N stage, microsatellite instability, perineural invasion), a nomogram was constructed. The C-index for the nomogram were 0.819 (95% CI=0.794-0.844), 0.782 (95% CI=0.740-0.824), 0.786 (95% CI=0.753-0.819), and 0.803 (95% CI=0.765-0.841) in the training set, internal validation set, external validation set 1, and external validation set 2, respectively. The calibration curves demonstrated a favorable congruence between the predicted and observed values as depicted by the nomogram. The decision curve analysis underscored that the nomogram yielded a heightened clinical net benefit.ConclusionThe constructed radiomics nomogram, amalgamating the radiomics features and clinical risk factors, exhibited commendable performance in the individualized prediction of postoperative DFS in CRC patients.
Collapse
Affiliation(s)
- Guodong Xu
- Department of Radiology, The Yancheng Clinical College of Xuzhou Medical University, The First People's Hospital of Yancheng, Yancheng, PR China
| | - Feng Feng
- Department of Radiology, Affiliated Tumor Hospital of Nantong University, Nantong, PR China
| | - Yanfen Cui
- Department of Radiology, Shanxi Cancer Hospital, Shanxi, PR China
| | - Yigang Fu
- Department of Radiology, The Yancheng Clinical College of Xuzhou Medical University, The First People's Hospital of Yancheng, Yancheng, PR China
| | - Yong Xiao
- Department of Radiology, The Yancheng Clinical College of Xuzhou Medical University, The First People's Hospital of Yancheng, Yancheng, PR China
| | - Wang Chen
- Department of Radiology, The Yancheng Clinical College of Xuzhou Medical University, The First People's Hospital of Yancheng, Yancheng, PR China
| | - Manman Li
- Department of Radiology, The Yancheng Clinical College of Xuzhou Medical University, The First People's Hospital of Yancheng, Yancheng, PR China
| |
Collapse
|
10
|
Delgadillo R, Spieler BO, Ford JC, Yang F, Studenski M, Padgett KR, Deana AM, Jin W, Abramowitz MC, Dal Pra A, Stoyanova R, Dogan N. Correlation of T2-Weighted Magnetic Resonance Imaging (MRI), Computed Tomography (CT), and Cone Beam Computed Tomography (CBCT) Radiomic Features for Prostate Cancer. Cureus 2025; 17:e80090. [PMID: 40190983 PMCID: PMC11970571 DOI: 10.7759/cureus.80090] [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] [Accepted: 02/20/2025] [Indexed: 04/09/2025] Open
Abstract
Radiomics extracted from cone beam computed tomography (CBCT) can be assessed at time points during treatment and may provide an advantage over assessments in a pre-treatment setting using diagnostic images, like magnetic resonance imaging (MRI) or computed tomography (CT), for prostate cancer (pCa) patients receiving radiotherapy (RT). The purpose of this study was to analyze correlations between prostate radiomic features (RFs) derived from T2-weighted (T2w) MRI, CT, and first fraction CBCT for patients receiving RT for pCa. Forty-seven patients were analyzed. The prostate volumes were manually segmented, and 42 radiomic features were extracted, of which seven volume-normalized RFs were considered. The absolute Spearman correlation was calculated among the RFs of the aforementioned imaging modalities (RM) and prostate volume (RV) since the motivation of this paper was to analyze the strength of the correlation. The Benjamini-Hochberg adjustment was applied to p-values to account for multiple comparisons. No high correlations were found between CT/CBCT vs. T2w. The intramodality RM demonstrated that CT RFs were much higher than the other modalities. For example, intramodality RM≥0.95 the percentage of RFs was 17% for CT, 9% for CBCT, and 4.5% for T2w. The differences in RFs across different modalities can be viewed positively: the lack of correlation between RFs across T2w and CT/CBCT could indicate a fundamental difference in the extractable image information. It could also indicate that some RFs did not have any extractable information. A future study will include evaluating the predictive performance of patient outcomes using radiomic features from CT, CBCT, and T2w, which could help in answering such questions.
Collapse
Affiliation(s)
- Rodrigo Delgadillo
- Radiation Oncology, University of Miami Sylvester Comprehensive Cancer Center, Miller School of Medicine, Miami, USA
| | - Benjamin O Spieler
- Radiation Oncology, University of Miami Sylvester Comprehensive Cancer Center, Miller School of Medicine, Miami, USA
| | - John C Ford
- Radiation Oncology, University of Miami Sylvester Comprehensive Cancer Center, Miller School of Medicine, Miami, USA
| | - Fei Yang
- Radiation Oncology, University of Miami Sylvester Comprehensive Cancer Center, Miller School of Medicine, Miami, USA
| | - Matthew Studenski
- Radiation Oncology, University of Miami Sylvester Comprehensive Cancer Center, Miller School of Medicine, Miami, USA
| | - Kyle R Padgett
- Radiation Oncology, University of Miami Sylvester Comprehensive Cancer Center, Miller School of Medicine, Miami, USA
| | - Anthony M Deana
- Radiation Oncology, Varian Medical Systems, Phoenix, USA
- Radiation Oncology, University of Miami Sylvester Comprehensive Cancer Center, Miller School of Medicine, Miami, USA
| | - William Jin
- Radiation Oncology, University of Miami Sylvester Comprehensive Cancer Center, Miller School of Medicine, Miami, USA
| | - Matthew C Abramowitz
- Radiation Oncology, University of Miami Sylvester Comprehensive Cancer Center, Miller School of Medicine, Miami, USA
| | - Alan Dal Pra
- Radiation Oncology, University of Miami Sylvester Comprehensive Cancer Center, Miller School of Medicine, Miami, USA
| | - Radka Stoyanova
- Radiation Oncology, University of Miami Sylvester Comprehensive Cancer Center, Miller School of Medicine, Miami, USA
| | - Nesrin Dogan
- Radiation Oncology, University of Miami Sylvester Comprehensive Cancer Center, Miller School of Medicine, Miami, USA
| |
Collapse
|
11
|
Widaatalla Y, Wolswijk T, Khan MD, Halilaj I, Mosterd K, Woodruff HC, Lambin P. Radiomics in Dermatological Optical Coherence Tomography (OCT): Feature Repeatability, Reproducibility, and Integration into Diagnostic Models in a Prospective Study. Cancers (Basel) 2025; 17:768. [PMID: 40075619 PMCID: PMC11899706 DOI: 10.3390/cancers17050768] [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: 01/24/2025] [Revised: 02/14/2025] [Accepted: 02/17/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND/OBJECTIVES Radiomics has seen substantial growth in medical imaging; however, its potential in optical coherence tomography (OCT) has not been widely explored. We systematically evaluate the repeatability and reproducibility of handcrafted radiomics features (HRFs) from OCT scans of benign nevi and examine the impact of bin width (BW) selection on HRF stability. The effect of using stable features on a radiomics classification model was also assessed. METHODS In this prospective study, 20 volunteers underwent test-retest OCT imaging of 40 benign nevi, resulting in 80 scans. The repeatability and reproducibility of HRFs extracted from manually delineated regions of interest (ROIs) were assessed using concordance correlation coefficients (CCCs) across BWs ranging from 5 to 50. A unique set of stable HRFs was identified at each BW after removing highly correlated features to eliminate redundancy. These robust features were incorporated into a multiclass radiomics classifier trained to distinguish benign nevi, basal cell carcinoma (BCC), and Bowen's disease. RESULTS Six stable HRFs were identified across all BWs, with a BW of 25 emerging as the optimal choice, balancing repeatability and the ability to capture meaningful textural details. Additionally, intermediate BWs (20-25) yielded 53 reproducible features. A classifier trained with six stable features achieved a 90% accuracy and AUCs of 0.96 and 0.94 for BCC and Bowen's disease, respectively, compared to a 76% accuracy and AUCs of 0.86 and 0.80 for a conventional feature selection approach. CONCLUSIONS This study highlights the critical role of BW selection in enhancing HRF stability and provides a methodological framework for optimizing preprocessing in OCT radiomics. By demonstrating the integration of stable HRFs into diagnostic models, we establish OCT radiomics as a promising tool to aid non-invasive diagnosis in dermatology.
Collapse
Affiliation(s)
- Yousif Widaatalla
- The D-Lab, Department of Precision Medicine, Maastricht University, 6200 MD Maastricht, The Netherlands; (M.D.K.); (I.H.); (H.C.W.); (P.L.)
- GROW Research Institute for Oncology and Reproduction, Maastricht University, 6200 MD Maastricht, The Netherlands; (T.W.); (K.M.)
| | - Tom Wolswijk
- GROW Research Institute for Oncology and Reproduction, Maastricht University, 6200 MD Maastricht, The Netherlands; (T.W.); (K.M.)
- Department of Dermatology, Maastricht University Medical Center+, 6229 HX Maastricht, The Netherlands
| | - Muhammad Danial Khan
- The D-Lab, Department of Precision Medicine, Maastricht University, 6200 MD Maastricht, The Netherlands; (M.D.K.); (I.H.); (H.C.W.); (P.L.)
- GROW Research Institute for Oncology and Reproduction, Maastricht University, 6200 MD Maastricht, The Netherlands; (T.W.); (K.M.)
| | - Iva Halilaj
- The D-Lab, Department of Precision Medicine, Maastricht University, 6200 MD Maastricht, The Netherlands; (M.D.K.); (I.H.); (H.C.W.); (P.L.)
- GROW Research Institute for Oncology and Reproduction, Maastricht University, 6200 MD Maastricht, The Netherlands; (T.W.); (K.M.)
| | - Klara Mosterd
- GROW Research Institute for Oncology and Reproduction, Maastricht University, 6200 MD Maastricht, The Netherlands; (T.W.); (K.M.)
- Department of Dermatology, Maastricht University Medical Center+, 6229 HX Maastricht, The Netherlands
| | - Henry C. Woodruff
- The D-Lab, Department of Precision Medicine, Maastricht University, 6200 MD Maastricht, The Netherlands; (M.D.K.); (I.H.); (H.C.W.); (P.L.)
- GROW Research Institute for Oncology and Reproduction, Maastricht University, 6200 MD Maastricht, The Netherlands; (T.W.); (K.M.)
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center+, 6229 HX Maastricht, The Netherlands
| | - Philippe Lambin
- The D-Lab, Department of Precision Medicine, Maastricht University, 6200 MD Maastricht, The Netherlands; (M.D.K.); (I.H.); (H.C.W.); (P.L.)
- GROW Research Institute for Oncology and Reproduction, Maastricht University, 6200 MD Maastricht, The Netherlands; (T.W.); (K.M.)
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center+, 6229 HX Maastricht, The Netherlands
| |
Collapse
|
12
|
Gennaro N, Soliman M, Borhani AA, Kelahan L, Savas H, Avery R, Subedi K, Trabzonlu TA, Krumpelman C, Yaghmai V, Chae Y, Lorch J, Mahalingam D, Mulcahy M, Benson A, Bagci U, Velichko YS. Delta Radiomics and Tumor Size: A New Predictive Radiomics Model for Chemotherapy Response in Liver Metastases from Breast and Colorectal Cancer. Tomography 2025; 11:20. [PMID: 40137560 PMCID: PMC11945686 DOI: 10.3390/tomography11030020] [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: 12/23/2024] [Revised: 02/14/2025] [Accepted: 02/18/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: Radiomic features exhibit a correlation with tumor size on pretreatment images. However, on post-treatment images, this association is influenced by treatment efficacy and varies between responders and non-responders. This study introduces a novel model, called baseline-referenced Delta radiomics, which integrates the association between radiomic features and tumor size into Delta radiomics to predict chemotherapy response in liver metastases from breast cancer (BC) and colorectal cancer (CRC). Materials and Methods: A retrospective study analyzed contrast-enhanced computed tomography (CT) scans of 83 BC patients and 84 CRC patients. Among these, 57 BC patients with 106 liver lesions and 37 CRC patients with 109 lesions underwent post-treatment imaging after systemic chemotherapy. Radiomic features were extracted from up to three lesions per patient following manual segmentation. Tumor response was assessed by measuring the longest diameter and classified according to RECIST 1.1 criteria as progressive disease (PD), partial response (PR), or stable disease (SD). Classification models were developed to predict chemotherapy response using pretreatment data only, Delta radiomics, and baseline-referenced Delta radiomics. Model performance was evaluated using confusion matrix metrics. Results: Baseline-referenced Delta radiomics performed comparably or better than established radiomics models in predicting tumor response in chemotherapy-treated patients with liver metastases. The sensitivity, specificity, and balanced accuracy in predicting response ranged from 0.66 to 0.97, 0.81 to 0.97, and 80% to 90%, respectively. Conclusions: By integrating the relationship between radiomic features and tumor size into Delta radiomics, baseline-referenced Delta radiomics offers a promising approach for predicting chemotherapy response in liver metastases from breast and colorectal cancer.
Collapse
Affiliation(s)
- Nicolò Gennaro
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (N.G.); (M.S.); (A.A.B.); (L.K.); (H.S.); (R.A.); (K.S.); (T.A.T.); (C.K.); (U.B.)
| | - Moataz Soliman
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (N.G.); (M.S.); (A.A.B.); (L.K.); (H.S.); (R.A.); (K.S.); (T.A.T.); (C.K.); (U.B.)
| | - Amir A. Borhani
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (N.G.); (M.S.); (A.A.B.); (L.K.); (H.S.); (R.A.); (K.S.); (T.A.T.); (C.K.); (U.B.)
| | - Linda Kelahan
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (N.G.); (M.S.); (A.A.B.); (L.K.); (H.S.); (R.A.); (K.S.); (T.A.T.); (C.K.); (U.B.)
| | - Hatice Savas
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (N.G.); (M.S.); (A.A.B.); (L.K.); (H.S.); (R.A.); (K.S.); (T.A.T.); (C.K.); (U.B.)
| | - Ryan Avery
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (N.G.); (M.S.); (A.A.B.); (L.K.); (H.S.); (R.A.); (K.S.); (T.A.T.); (C.K.); (U.B.)
| | - Kamal Subedi
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (N.G.); (M.S.); (A.A.B.); (L.K.); (H.S.); (R.A.); (K.S.); (T.A.T.); (C.K.); (U.B.)
| | - Tugce A. Trabzonlu
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (N.G.); (M.S.); (A.A.B.); (L.K.); (H.S.); (R.A.); (K.S.); (T.A.T.); (C.K.); (U.B.)
| | - Chase Krumpelman
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (N.G.); (M.S.); (A.A.B.); (L.K.); (H.S.); (R.A.); (K.S.); (T.A.T.); (C.K.); (U.B.)
| | - Vahid Yaghmai
- Department of Radiological Sciences, University of California Irvine, Irvine, CA 92868, USA;
| | - Young Chae
- Department of Medicine, Division of Hematology/Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (Y.C.); (J.L.); (D.M.); (M.M.); (A.B.)
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA
| | - Jochen Lorch
- Department of Medicine, Division of Hematology/Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (Y.C.); (J.L.); (D.M.); (M.M.); (A.B.)
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA
| | - Devalingam Mahalingam
- Department of Medicine, Division of Hematology/Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (Y.C.); (J.L.); (D.M.); (M.M.); (A.B.)
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA
| | - Mary Mulcahy
- Department of Medicine, Division of Hematology/Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (Y.C.); (J.L.); (D.M.); (M.M.); (A.B.)
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA
| | - Al Benson
- Department of Medicine, Division of Hematology/Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (Y.C.); (J.L.); (D.M.); (M.M.); (A.B.)
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA
| | - Ulas Bagci
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (N.G.); (M.S.); (A.A.B.); (L.K.); (H.S.); (R.A.); (K.S.); (T.A.T.); (C.K.); (U.B.)
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA
| | - Yuri S. Velichko
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (N.G.); (M.S.); (A.A.B.); (L.K.); (H.S.); (R.A.); (K.S.); (T.A.T.); (C.K.); (U.B.)
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
13
|
Oh G, Gi Y, Lee J, Kim H, Wu HG, Park JM, Choi E, Shin D, Yoon M, Lee B, Son J. Hybrid Approach to Classifying Histological Subtypes of Non-small Cell Lung Cancer (NSCLC): Combining Radiomics and Deep Learning Features from CT Images. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2025:10.1007/s10278-025-01442-5. [PMID: 39953259 DOI: 10.1007/s10278-025-01442-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 01/15/2025] [Accepted: 02/04/2025] [Indexed: 02/17/2025]
Abstract
This study aimed to develop a hybrid model combining radiomics and deep learning features derived from computed tomography (CT) images to classify histological subtypes of non-small cell lung cancer (NSCLC). We analyzed CT images and radiomics features from 235 patients with NSCLC, including 110 with adenocarcinoma (ADC) and 112 with squamous cell carcinoma (SCC). The dataset was split into a training set (75%) and a test set (25%). External validation was conducted using the NSCLC-Radiomics database, comprising 24 patients each with ADC and SCC. A total of 1409 radiomics and 8192 deep features underwent principal component analysis (PCA) and ℓ2,1-norm minimization for feature reduction and selection. The optimal feature sets for classification included 27 radiomics features, 20 deep features, and 55 combined features (30 deep and 25 radiomics). The average area under the receiver operating characteristic curve (AUC) for radiomics, deep, and combined features were 0.6568, 0.6689, and 0.7209, respectively, across the internal and external test sets. Corresponding average accuracies were 0.6013, 0.6376, and 0.6564. The combined model demonstrated superior performance in classifying NSCLC subtypes, achieving higher AUC and accuracy in both test datasets. These results suggest that the proposed hybrid approach could enhance the accuracy and reliability of NSCLC subtype classification.
Collapse
Affiliation(s)
- Geon Oh
- Department of Bioengineering, Korea University, Seoul, Republic of Korea
- Proton Therapy Center, National Cancer Center, Goyang, Republic of Korea
| | - Yongha Gi
- Department of Bioengineering, Korea University, Seoul, Republic of Korea
| | - Jeongshim Lee
- Department of Radiation Oncology, Inha University Hospital, 27, Inhang-Ro, Jung-Gu, Incheon, 22332, Republic of Korea
| | - Hunjung Kim
- Department of Radiation Oncology, Inha University Hospital, 27, Inhang-Ro, Jung-Gu, Incheon, 22332, Republic of Korea
| | - Hong-Gyun Wu
- Department of Radiation Oncology, Seoul National University Hospital, 101, Daehak-Ro, Jongno-Gu, Seoul, Republic of Korea
| | - Jong Min Park
- Department of Radiation Oncology, Seoul National University Hospital, 101, Daehak-Ro, Jongno-Gu, Seoul, Republic of Korea
| | - Eunae Choi
- Department of Radiological Science, Daegu Catholic University, Gyeongsan, Gyeongsangbuk-Do, Korea
| | - Dongho Shin
- Proton Therapy Center, National Cancer Center, Goyang, Republic of Korea
| | - Myonggeun Yoon
- Department of Bioengineering, Korea University, Seoul, Republic of Korea
| | - Boram Lee
- Department of Radiation Oncology, Inha University Hospital, 27, Inhang-Ro, Jung-Gu, Incheon, 22332, Republic of Korea.
| | - Jaeman Son
- Department of Radiation Oncology, Seoul National University Hospital, 101, Daehak-Ro, Jongno-Gu, Seoul, Republic of Korea.
| |
Collapse
|
14
|
Wang H, Hong Y, Zhang Z, Cheng K, Chen B, Zhang R. Study on postoperative survival prediction model for non-small cell lung cancer: application of radiomics technology workflow based on multi-organ imaging features and various machine learning algorithms. Front Med (Lausanne) 2025; 12:1517765. [PMID: 39975681 PMCID: PMC11835680 DOI: 10.3389/fmed.2025.1517765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 01/24/2025] [Indexed: 02/21/2025] Open
Abstract
Objective This study aims to construct an effective prediction model for the two-year postoperative survival probability of patients with non-small cell lung cancer (NSCLC). It particularly focuses on integrating radiomics features, including the erector spinae and whole-lung imaging features, to enhance the accuracy and stability of prognostic predictions. Materials and methods The study included 37 NSCLC patients diagnosed and surgically treated at the First Affiliated Hospital of Anhui Medical University from January 2020 to December 2021. The average age of the patients was 59 years, with the majority being female and non-smokers. Additionally, CT imaging data from 98 patients were obtained from The Cancer Imaging Archive (TCIA) public database. All imaging data were derived from preoperative chest CT scans and standardized using 3D Slicer software. The study extracted radiomic features from the tumor, whole lung, and erector spinae muscles of the patients and applied 11 machine learning algorithms to construct prediction models. Subsequently, the classification performance of all constructed models was compared to select the optimal prediction model. Results Univariate Cox regression analysis showed no significant correlation between the collected clinical factors and patient survival time. In the external validation set, the K-Nearest Neighbors (KNN) model based on bilateral erector spinae features performed the best, with accuracy and AUC (Area Under the Curve) values consistently above 0.7 in both the training and external testing sets. Among the prognostic models based on whole-lung imaging features, the AdaBoost model also performed well, but its AUC value was below 0.6 in the external validation set, indicating overall classification performance still inferior to the KNN model based on erector spinae features. Conclusion This study is the first to introduce erector spinae imaging features into lung cancer research, successfully developing a stable and well-performing prediction model for the postoperative survival of NSCLC patients. The research results provide new perspectives and directions for the application of radiomics in cancer research and emphasize the importance of incorporating multi-organ imaging features to improve the accuracy and stability of prediction models.
Collapse
Affiliation(s)
- Hanlin Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yuan Hong
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zimo Zhang
- Department of The First Clinical Medical College, Anhui Medical University, Hefei, China
| | - Kang Cheng
- Department of The First Clinical Medical College, Anhui Medical University, Hefei, China
| | - Bo Chen
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Renquan Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
15
|
Fusco R, Granata V, Setola SV, Trovato P, Galdiero R, Mattace Raso M, Maio F, Porto A, Pariante P, Cerciello V, Sorgente E, Pecori B, Castaldo M, Izzo F, Petrillo A. The application of radiomics in cancer imaging with a focus on lung cancer, renal cell carcinoma, gastrointestinal cancer, and head and neck cancer: A systematic review. Phys Med 2025; 130:104891. [PMID: 39787678 DOI: 10.1016/j.ejmp.2025.104891] [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: 06/08/2024] [Revised: 12/31/2024] [Accepted: 01/02/2025] [Indexed: 01/12/2025] Open
Abstract
PURPOSE To study the application of radiomics in cancer imaging with a focus on lung cancer, renal cell carcinoma, gastrointestinal cancer, and head and neck cancer. METHODS Different electronic databases were considered. Articles published in the last five years were analyzed (January 2019 and December 2023). Papers were selected by two investigators with over 15 years of experience in Radiomics analysis in cancer imaging. The methodological quality of each radiomics study was performed using the Radiomic Quality Score (RQS) by two different readers in consensus and then by a third operator to solve disagreements between the two readers. RESULTS 19 articles are included in the review. Among the analyzed studies, only one study achieved an RQS of 18 reporting multivariable analyzes with also non-radiomics features and using the validation phase considering two datasets from two distinct institutes and open science and data domain. CONCLUSION This informative review has brought attention to the increasingly consolidated potential of Radiomics, although there are still several aspects to be evaluated before the transition to routine clinical practice. There are several challenges to address, including the need for standardization at all stages of the workflow and the potential for cross-site validation using heterogeneous real-world datasets. It will be necessary to establish and promote an imaging data acquisition protocol, conduct multicenter prospective quality control studies, add scanner differences and vendor-dependent characteristics; to collect images of individuals at additional time points, to report calibration statistics.
Collapse
Affiliation(s)
- Roberta Fusco
- Division of Radiology, "Istituto Nazionale Tumori IRCCS Fondazione Pascale, IRCCS di Napoli, 80131 Naples, Italy
| | - Vincenza Granata
- Division of Radiology, "Istituto Nazionale Tumori IRCCS Fondazione Pascale, IRCCS di Napoli, 80131 Naples, Italy.
| | - Sergio Venanzio Setola
- Division of Radiology, "Istituto Nazionale Tumori IRCCS Fondazione Pascale, IRCCS di Napoli, 80131 Naples, Italy
| | - Piero Trovato
- Division of Radiology, "Istituto Nazionale Tumori IRCCS Fondazione Pascale, IRCCS di Napoli, 80131 Naples, Italy
| | - Roberta Galdiero
- Division of Radiology, "Istituto Nazionale Tumori IRCCS Fondazione Pascale, IRCCS di Napoli, 80131 Naples, Italy
| | - Mauro Mattace Raso
- Division of Radiology, "Istituto Nazionale Tumori IRCCS Fondazione Pascale, IRCCS di Napoli, 80131 Naples, Italy
| | - Francesca Maio
- Division of Radiology, "Istituto Nazionale Tumori IRCCS Fondazione Pascale, IRCCS di Napoli, 80131 Naples, Italy
| | - Annamaria Porto
- Division of Radiology, "Istituto Nazionale Tumori IRCCS Fondazione Pascale, IRCCS di Napoli, 80131 Naples, Italy
| | - Paolo Pariante
- Division of Radiology, "Istituto Nazionale Tumori IRCCS Fondazione Pascale, IRCCS di Napoli, 80131 Naples, Italy
| | - Vincenzo Cerciello
- Division of Health Physics, "Istituto Nazionale Tumori IRCCS Fondazione Pascale, IRCCS di Napoli, 80131 Naples, Italy
| | - Eugenio Sorgente
- Division of Radiation protection and innovative technology, Istituto Nazionale Tumori IRCCS Fondazione Pascale, IRCCS di Napoli, 80131 Naples, Italy
| | - Biagio Pecori
- Division of Radiation protection and innovative technology, Istituto Nazionale Tumori IRCCS Fondazione Pascale, IRCCS di Napoli, 80131 Naples, Italy
| | - Mimma Castaldo
- Unit of "Progettazione e Manutenzione Edile ed impianti", Istituto Nazionale Tumori IRCCS Fondazione Pascale, IRCCS di Napoli, 80131 Naples, Italy
| | - Francesco Izzo
- Division of Epatobiliary Surgical Oncology, Istituto Nazionale Tumori IRCCS Fondazione Pascale, IRCCS di Napoli, 80131 Naples, Italy
| | - Antonella Petrillo
- Division of Radiology, "Istituto Nazionale Tumori IRCCS Fondazione Pascale, IRCCS di Napoli, 80131 Naples, Italy
| |
Collapse
|
16
|
Kravchenko D, Vecsey-Nagy M, Varga-Szemes A, Hagar MT, Schoepf UJ, Gnasso C, Zsarnóczay E, O'Doherty J, Caruso D, Laghi A, Emrich T, Tremamunno G. Intra-individual radiomic analysis of pericoronary adipose tissue: Photon-counting detector vs energy-integrating detector CT angiography. Int J Cardiol 2025; 420:132749. [PMID: 39579791 DOI: 10.1016/j.ijcard.2024.132749] [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: 09/17/2024] [Revised: 11/12/2024] [Accepted: 11/18/2024] [Indexed: 11/25/2024]
Abstract
BACKGROUND The impact of novel photon-counting detector (PCD)-CT technology on in-vivo radiomics is not fully understood. This study aimed to compare the intra-individual stability and reproducibility of pericoronary adipose tissue (PCAT) radiomic features between PCD-CT and energy-integrating detector (EID)-CT in patients undergoing coronary CT angiography (CCTA) on both systems. METHODS Patients undergoing clinically indicated CCTA on an EID-CT were prospectively enrolled for research PCD-CCTA within 30 days. Image acquisition parameters were standardized; PCD-CT datasets were reconstructed both down-sampled to 0.6 mm to match the clinical scan (PCD-CTDS) and at 0.2 mm ultrahigh-resolution mode (PCD-CTUHR). Automatic PCAT segmentation was performed; a total of 110 radiomic feature classes were extracted and compared across the three datasets (EID-CT, PCD-CTDS, and PCD-CDUHR). Feature stability was assessed using paired t-test filtered for false discoveries using Benjamini-Hochberg method, and reproducibility using intraclass correlation coefficient (ICC). RESULTS A total of 42 patients (34 male [81.0 %]; 67.9 ± 7.6 years) were included. Feature stability was 91 % for EID-CT vs. PCD-CTDS, but decreased for UHR datasets (EID-CT vs. PCD-CTUHR: 55 %; PCD-CTDS vs. PCD-CTUHR: 51 %). However, inter-scanner reproducibility was poor in both comparisons (EID-CT vs. PCD-CTDS median ICC: 0.43 [0.03-0.69]; EID-CT vs. PCD-CTUHR: 0.29 [0.01-0.51]). Nevertheless, reproducibility improved within PCD-CT datasets (PCD-CTDS vs. PCD-CTUHR: 0.72 [0.48-0.83]), regardless of the difference in slice thickness. CONCLUSIONS Most PCAT radiomic features remained stable between EID-CT and PCD-CTDS, although inter-scanner reproducibility was poor, emphasizing the significant impact of detector technology. Conversely, reproducibility of features within PCD-CT datasets showed more consistent results, even when comparing standard to UHR.
Collapse
Affiliation(s)
- Dmitrij Kravchenko
- Department of Radiology and Radiological Science, Medical University of South Carolina, Charleston, SC, USA; Department of Diagnostic and Interventional Radiology, University Hospital Bonn, Bonn, Germany; Quantitative Imaging Laboratory Bonn (QILaB), Bonn, Germany.
| | - Milan Vecsey-Nagy
- Department of Radiology and Radiological Science, Medical University of South Carolina, Charleston, SC, USA; Heart and Vascular Centre, Semmelweis University, Budapest, Hungary.
| | - Akos Varga-Szemes
- Department of Radiology and Radiological Science, Medical University of South Carolina, Charleston, SC, USA.
| | - Muhammad Taha Hagar
- Department of Radiology and Radiological Science, Medical University of South Carolina, Charleston, SC, USA; Department of Diagnostic and Interventional Radiology, Medical Center - University of Freiburg, Freiburg, Germany.
| | - U Joseph Schoepf
- Department of Radiology and Radiological Science, Medical University of South Carolina, Charleston, SC, USA.
| | - Chiara Gnasso
- Department of Radiology and Radiological Science, Medical University of South Carolina, Charleston, SC, USA; Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| | - Emese Zsarnóczay
- Department of Radiology and Radiological Science, Medical University of South Carolina, Charleston, SC, USA; MTA-SE Cardiovascular Imaging Research Group, Department of Radiology, Medical Imaging Centre, Semmelweis University, Hungary.
| | - Jim O'Doherty
- Department of Radiology and Radiological Science, Medical University of South Carolina, Charleston, SC, USA; Siemens Medical Solutions, Malvern, PA, USA.
| | - Damiano Caruso
- Department of Medical Surgical Sciences and Translational Medicine, Sapienza University of Rome, Italy.
| | - Andrea Laghi
- Department of Medical Surgical Sciences and Translational Medicine, Sapienza University of Rome, Italy.
| | - Tilman Emrich
- Department of Radiology and Radiological Science, Medical University of South Carolina, Charleston, SC, USA; Department of Diagnostic and Interventional Radiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner-Site Rhine-Main, Mainz, Germany.
| | - Giuseppe Tremamunno
- Department of Radiology and Radiological Science, Medical University of South Carolina, Charleston, SC, USA; Department of Medical Surgical Sciences and Translational Medicine, Sapienza University of Rome, Italy.
| |
Collapse
|
17
|
Anderson P, Dogan N, Ford JC, Padgett K, Simpson G, Stoyanova R, Abramowitz MC, Dal Pra A, Delgadillo R. Repeatability, reproducibility, and the effects of radiotherapy on radiomic features of lowfield MR-LINAC images of the prostate. Front Oncol 2025; 14:1408752. [PMID: 39902123 PMCID: PMC11788350 DOI: 10.3389/fonc.2024.1408752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 12/24/2024] [Indexed: 02/05/2025] Open
Abstract
Definitive radiotherapy (RT) has been shown to be a successful method of treating prostate cancer (PCa) patients. Through radiomics, a quantitative analysis of medical images, it is possible to adapt treatment early on, which may prevent or mitigate future adverse events. During RT of PCa, low-field magnetic resonance (MR) images, taken with a LINAC onboard imaging system in a process known as magnetic resonance-guided radiotherapy (MRgRT), are used to improve treatment accuracy via superior setup compared to x-ray methods. This work investigated baseline repeatability of radiomic features (RFs) by comparing planning MR images (pMR) with first-fraction setup images (FX1) taken with onboard MRI. The changes in RFs following RT were also looked at with the use of last-fraction setup images (FX5). Earlier research has investigated the use of planning images from cone beam CT (CBCT), but to our knowledge no research has previously shown the relationship with onboard MRI. The correlation between FX1 images and 3T diagnostic MR (dT2) images was also studied. Forty-three first and second order radiomic features extracted from these images were compared by calculating Lin's concordance correlation coefficient (with Benjamini-Hochberg correction for multiple comparisons) between the modalities. FX1 and pMR images were correlated (p<0.05) for all but one RF. 12 RFs correlated between pMR and dT2 images. There was a noticeable change in correlation values for RFs when looking at FX1 and FX5 images, with only 15 correlating significantly. The change in correlation values between pMR and FX5 images was comparable to that between FX1 and FX5 images, with 33 features having a CCC value deviation of less than 0.1. These results demonstrate that RF features are repeatable across different images of the same modality without treatment intervention. This study has also shown a noticeable, reproducible change in RFs as RT goes on. Reproducibility of RFs between different modalities was not strong. This study demonstrated that we can reliably use onboard MRI to observe day-to-day feature changes as a result of RT.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Rodrigo Delgadillo
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
18
|
Ling X, Bazyar S, Ferris M, Molitoris J, Allor E, Thomas H, Arons D, Schumaker L, Krc R, Mendes WS, Tran PT, Sawant A, Mehra R, Gaykalova DA, Ren L. Identification of CT based radiomic biomarkers for progression free survival in head and neck squamous cell carcinoma. Sci Rep 2025; 15:1279. [PMID: 39779914 PMCID: PMC11711663 DOI: 10.1038/s41598-025-85498-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 01/03/2025] [Indexed: 01/11/2025] Open
Abstract
This study addresses the limited noninvasive tools for Head and Neck Squamous Cell Carcinoma (HNSCC) progression-free survival (PFS) prediction by identifying Computed Tomography (CT)-based biomarkers for predicting prognosis. A retrospective analysis was conducted on data from 203 HNSCC patients. An ensemble feature selection involving correlation analysis, univariate survival analysis, best-subset selection, and the LASSO-Cox algorithm was used to select functional features, which were then used to build final Cox Proportional Hazards models (CPH). Our CPH achieved a 0.69 concordance index in an external indepedent cohort of 77 patients. The model identified five CT-based radiomics features, Gradient ngtdm Contrast, Logσ=33D-FirstorderRootMeanSquared, Logσ=0.13D-glszm SmallAreaLowGrayLevelEmphasis, Exponential-gldm LargeDependenceHighGrayLevelEmphasis, and Gradient ngtdm Strength as survival biomarkers (p-value < 0.05). These findings contribute to our knowledge of how radiomics can be used to predict the outcome so that treatment plans can be tailored for people with HNSCC to improve their prognosis.
Collapse
Affiliation(s)
- Xiao Ling
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
- Mathematics Department, Auburn University at Montgomery, Alabama, USA
| | - Soha Bazyar
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Matthew Ferris
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jason Molitoris
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Erin Allor
- University of Maryland School of Medicine, Baltimore, MD, USA
| | - Hannah Thomas
- University of Maryland School of Medicine, Baltimore, MD, USA
| | - Danielle Arons
- University of Maryland School of Medicine, Baltimore, MD, USA
| | - Lisa Schumaker
- University of Maryland School of Medicine, Baltimore, MD, USA
| | - Rebecca Krc
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - William Silva Mendes
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Phuoc T Tran
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Amit Sawant
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Ranee Mehra
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Daria A Gaykalova
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Otorhinolaryngology-Head and Neck Surgery, Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, Baltimore, MD, USA
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Lei Ren
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
19
|
Magnin CY, Lauer D, Ammeter M, Gote-Schniering J. From images to clinical insights: an educational review on radiomics in lung diseases. Breathe (Sheff) 2025; 21:230225. [PMID: 40104259 PMCID: PMC11915127 DOI: 10.1183/20734735.0225-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 12/16/2024] [Indexed: 03/20/2025] Open
Abstract
Radiological imaging is a cornerstone in the clinical workup of lung diseases. Radiomics represents a significant advancement in clinical lung imaging, offering a powerful tool to complement traditional qualitative image analysis. Radiomic features are quantitative and computationally describe shape, intensity, texture and wavelet characteristics from medical images that can uncover detailed and often subtle information that goes beyond the visual capabilities of radiological examiners. By extracting this quantitative information, radiomics can provide deep insights into the pathophysiology of lung diseases and support clinical decision-making as well as personalised medicine approaches. In this educational review, we provide a step-by-step guide to radiomics-based medical image analysis, discussing the technical challenges and pitfalls, and outline the potential clinical applications of radiomics in diagnosing, prognosticating and evaluating treatment responses in respiratory medicine.
Collapse
Affiliation(s)
- Cheryl Y Magnin
- Department of Rheumatology and Immunology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Both authors contributed equally
| | - David Lauer
- Department of Rheumatology and Immunology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Both authors contributed equally
| | - Michael Ammeter
- Department of Rheumatology and Immunology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Janine Gote-Schniering
- Department of Rheumatology and Immunology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department of Pulmonary Medicine, Allergology and Clinical Immunology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
20
|
Liu L, Zhang W, Wang Y, Wu J, Fan Q, Chen W, Zhou L, Li J, Li Y. Radiomics combined with clinical and MRI features may provide preoperative evaluation of suboptimal debulking surgery for serous ovarian carcinoma. Abdom Radiol (NY) 2025; 50:496-512. [PMID: 39003651 PMCID: PMC11711150 DOI: 10.1007/s00261-024-04343-3] [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: 02/27/2024] [Revised: 04/10/2024] [Accepted: 04/16/2024] [Indexed: 07/15/2024]
Abstract
PURPOSE To develop and validate a model for predicting suboptimal debulking surgery (SDS) of serous ovarian carcinoma (SOC) using radiomics method, clinical and MRI features. METHODS 228 patients eligible from institution A (randomly divided into the training and internal validation cohorts) and 45 patients from institution B (external validation cohort) were collected and retrospectively analyzed. All patients underwent abdominal pelvic enhanced MRI scan, including T2-weighted imaging fat-suppressed fast spin-echo (T2FSE), T1-weighted dual-echo magnetic resonance imaging (T1DEI), diffusion weighted imaging (DWI), and T1 with contrast enhancement (T1CE). We extracted, selected and eliminated highly correlated radiomic features for each sequence. Then, Radiomic models were made by each single sequence, dual-sequence (T1CE + T2FSE), and all-sequence, respectively. Univariate and multivariate analyses were performed to screen the clinical and MRI independent predictors. The radiomic model with the highest area under the curve (AUC) was used to combine the independent predictors as a combined model. RESULTS The optimal radiomic model was based on dual sequences (T2FSE + T1CE) among the five radiomic models (AUC = 0.720, P < 0.05). Serum carbohydrate antigen 125, the relationship between sigmoid colon/rectum and ovarian mass or mass implanted in Douglas' pouch, diaphragm nodules, and peritoneum/mesentery nodules were considered independent predictors. The AUC of the radiomic-clinical-radiological model was higher than either the optimal radiomic model or the clinical-radiological model in the training cohort (AUC = 0.908 vs. 0.720/0.854). CONCLUSIONS The radiomic-clinical-radiological model has an overall algorithm reproducibility and may help create individualized treatment programs and improve the prognosis of patients with SOC.
Collapse
Affiliation(s)
- Li Liu
- Department of Radiology, The People's Hospital of Yubei District of Chongqing City, No. 23 ZhongyangGongyuanBei Road, Yubei District, Chongqing, 401120, China
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuanjiagang, Yuzhong District, Chongqing, 400016, China
| | - Wenfei Zhang
- Department of Radiology, The People's Hospital of Yubei District of Chongqing City, No. 23 ZhongyangGongyuanBei Road, Yubei District, Chongqing, 401120, China
| | - Yudong Wang
- Institute of Clinical Algorithms, InferVision, Ocean International Center, Chaoyang District, Beijing, 100020, China
| | - Jiangfen Wu
- Institute of Clinical Algorithms, InferVision, Ocean International Center, Chaoyang District, Beijing, 100020, China
| | - Qianrui Fan
- Institute of Clinical Algorithms, InferVision, Ocean International Center, Chaoyang District, Beijing, 100020, China
| | - Weidao Chen
- Institute of Clinical Algorithms, InferVision, Ocean International Center, Chaoyang District, Beijing, 100020, China
| | - Linyi Zhou
- Department of Radiology, Daping Hospital, Army Medical Center, Army Medical University, 10# Changjiangzhilu, Chongqing, 40024, China
| | - Juncai Li
- Department of Surgery, The People's Hospital of Yubei District of Chongqing City, No. 23 ZhongyangGongyuanBei Road, Yubei District, Chongqing, 401120, China.
| | - Yongmei Li
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuanjiagang, Yuzhong District, Chongqing, 400016, China.
| |
Collapse
|
21
|
Bhandari A, Johnson K, Oh K, Yu F, Huynh LM, Lei Y, Wisnoskie S, Zhou S, Baine MJ, Lin C, Zhang C, Wang S. Developing a novel dosiomics model to predict treatment failures following lung stereotactic body radiation therapy. Front Oncol 2024; 14:1438861. [PMID: 39726705 PMCID: PMC11669717 DOI: 10.3389/fonc.2024.1438861] [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: 05/26/2024] [Accepted: 11/07/2024] [Indexed: 12/28/2024] Open
Abstract
Purpose The purpose of this study was to investigate the dosiomics features of the interplay between CT density and dose distribution in lung SBRT plans, and to develop a model to predict treatment failure following lung SBRT treatment. Methods A retrospective study was conducted involving 179 lung cancer patients treated with SBRT at the University of Nebraska Medical Center (UNMC) between October 2007 and June 2022. Features from the CT image, Biological Effective Dose (BED) and five interaction matrices between CT and BED were extracted using radiomics mathematics. Our in-house feature selection pipeline was utilized to evaluate and rank features based on their importance and redundancy, with only the selected non-redundant features being used for predictive modeling. We randomly selected 151 cases and 28 cases as training and test datasets. Four different models were trained utilizing the Balanced Random Forest framework on the same training dataset to differentiate between failure and non-failure cases. These four models utilized the same number of selected features extracted from CT-only, BED-only, a combination of CT and BED, and a composite of CT and BED including their interaction matrices, respectively. Results The cohort included 125 non-failure cases and 54 failure cases, with a median follow-up time of 34.4 months. We selected the top 17 important and non-redundant features (with the Pearsons's coefficient < 0.5) in each model. When evaluated on the same independent test set, the four models-CT features-only, BED features-only, a combination of CT and BED features, and a composite model including features from CT and BED that includes their interaction matrices-achieved AUC values of 0.56, 0.75, 0.73, and 0.82, respectively, with corresponding accuracies of 0.61, 0.79, 0.71, and 0.79. The composite model demonstrated the highest AUC and accuracy, indicating that incorporating interactions between CT and BED reveals more predictive capabilities in distinguishing between failure and non-failure cases. Conclusion The dosiomics model integrating the interaction between CT and Dose can effectively predict treatment failure following lung SBRT treatment and may serve as a useful tool to proactively evaluate and select lung SBRT treatment plans to reduce treatment failure in the future.
Collapse
Affiliation(s)
- Ashok Bhandari
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Kurtis Johnson
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Kyuhak Oh
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Fang Yu
- Department of Biostatistics, University of Nebraska Medical Center, Omaha, NE, United States
| | - Linda M. Huynh
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Yu Lei
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Sarah Wisnoskie
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE, United States
- Department of Radiation Oncology, Novant Health Cancer Institute, Winston-Salem, NC, United States
| | - Sumin Zhou
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Michael James Baine
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Chi Lin
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Chi Zhang
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Shuo Wang
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
22
|
Zhou T, Zhou X, Ni J, Guan Y, Jiang X, Lin X, Li J, Xia Y, Wang X, Wang Y, Huang W, Tu W, Dong P, Li Z, Liu S, Fan L. A CT-Based Lung Radiomics Nomogram for Classifying the Severity of Chronic Obstructive Pulmonary Disease. Int J Chron Obstruct Pulmon Dis 2024; 19:2705-2717. [PMID: 39677830 PMCID: PMC11646399 DOI: 10.2147/copd.s483007] [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: 08/16/2024] [Accepted: 12/02/2024] [Indexed: 12/17/2024] Open
Abstract
Background Chronic obstructive pulmonary disease (COPD) is a major global health concern, and while traditional pulmonary function tests are effective, recent radiomics advancements offer enhanced evaluation by providing detailed insights into the heterogeneous lung changes. Purpose To develop and validate a radiomics nomogram based on clinical and whole-lung computed tomography (CT) radiomics features to stratify COPD severity. Patients and Methods One thousand ninety-nine patients with COPD (including 308, 132, and 659 in the training, internal and external validation sets, respectively), confirmed by pulmonary function test, were enrolled from two institutions. The whole-lung radiomics features were obtained after a fully automated segmentation. Thereafter, a clinical model, radiomics signature, and radiomics nomogram incorporating radiomics signature as well as independent clinical factors were constructed and validated. Additionally, receiver-operating characteristic (ROC) curve, area under the ROC curve (AUC), decision curve analysis (DCA), and the DeLong test were used for performance assessment and comparison. Results In comparison with clinical model, both radiomics signature and radiomics nomogram outperformed better on COPD severity (GOLD I-II and GOLD III-IV) in three sets. The AUC of radiomics nomogram integrating age, height and Radscore, was 0.865 (95% CI, 0.818-0.913), 0.851 (95% CI, 0.778-0.923), and 0.781 (95% CI, 0.740-0.823) in three sets, which was the highest among three models (0.857; 0.850; 0.774, respectively) but not significantly different (P > 0.05). Decision curve analysis demonstrated the superiority of the radiomics nomogram in terms of clinical usefulness. Conclusion The present work constructed and verified the novel, diagnostic radiomics nomogram for identifying the severity of COPD, showing the added value of chest CT to evaluate not only the pulmonary structure but also the lung function status.
Collapse
Affiliation(s)
- Taohu Zhou
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, People’s Republic of China
- School of Medical Imaging, Shandong Second Medical University, Weifang, Shandong, People’s Republic of China
| | - Xiuxiu Zhou
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, People’s Republic of China
| | - Jiong Ni
- Department of Radiology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Yu Guan
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, People’s Republic of China
| | - Xin’ang Jiang
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, People’s Republic of China
| | - Xiaoqing Lin
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, People’s Republic of China
- College of Health Sciences and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, People’s Republic of China
| | - Jie Li
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, People’s Republic of China
- College of Health Sciences and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, People’s Republic of China
| | - Yi Xia
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, People’s Republic of China
| | - Xiang Wang
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, People’s Republic of China
| | - Yun Wang
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, People’s Republic of China
| | - Wenjun Huang
- Department of Radiology, The Second People’s Hospital of Deyang, Deyang, Sichuan, People’s Republic of China
| | - Wenting Tu
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, People’s Republic of China
| | - Peng Dong
- School of Medical Imaging, Shandong Second Medical University, Weifang, Shandong, People’s Republic of China
| | - Zhaobin Li
- Department of Radiation Oncology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, 200233, People’s Republic of China
| | - Shiyuan Liu
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, People’s Republic of China
| | - Li Fan
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, People’s Republic of China
| |
Collapse
|
23
|
Ou J, Zhou HY, Qin HL, Wang YS, Gou YQ, Luo H, Zhang XM, Chen TW. Baseline CT radiomics features to predict pathological complete response of advanced esophageal squamous cell carcinoma treated with neoadjuvant chemotherapy using paclitaxel and cisplatin. Eur J Radiol 2024; 181:111763. [PMID: 39341168 DOI: 10.1016/j.ejrad.2024.111763] [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: 07/02/2024] [Revised: 09/20/2024] [Accepted: 09/25/2024] [Indexed: 09/30/2024]
Abstract
PURPOSE To develop a CT radiomics model to predict pathological complete response (pCR) of advanced esophageal squamous cell carcinoma (ESCC) toneoadjuvant chemotherapy using paclitaxel and cisplatin. MATERIALS AND METHODS 326 consecutive patients with advanced ESCC from two hospitals undergoing baseline contrast-enhanced CT followed by neoadjuvant chemotherapy using paclitaxel and cisplatin were enrolled, including 115 patients achieving pCR and 211 patients without pCR. Of the 271 cases from 1st hospital, 188 and 83 cases were randomly allocated to the training and test cohorts, respectively. The 55 patients from a second hospital were assigned as an external validation cohort. Region of interest was segmented on the baseline thoracic contrast-enhanced CT. Useful radiomics features were generated by dimension reduction using least absolute shrinkage and selection operator. The optimal radiomics features were chosen using support vector machine (SVM). Discriminating performance was assessed with area under the receiver operating characteristic curve (ROC) and F-1score. The calibration curves and Brier score were used to evaluate the predictive accuracy. RESULTS Eight radiomics features were selected to create radiomics models related to pCR of advanced ESCC (P-values < 0.01 for both the training and test cohorts). SVM model showed the best performance (AUCs = 0.929, 0.868 and 0.866, F-1scores = 0.857, 0.847 and 0.737 in the training, test and external validation cohorts, respectively). The calibration curves and Brier scores indicated goodness-of-fit and its great predictive accuracy. CONCLUSION CT radiomics models could well help predict pCR of advanced ESCC, and SVM model could be a suitable predictive model.
Collapse
Affiliation(s)
- Jing Ou
- The First Clinical College of Jinan University, and Jinan University First Affiliated Hospital, Guangzhou, Guangdong 510630, China; Medical Imaging Key Laboratory of Sichuan Province, and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China
| | - Hai-Ying Zhou
- The First Clinical College of Jinan University, and Jinan University First Affiliated Hospital, Guangzhou, Guangdong 510630, China; Medical Imaging Key Laboratory of Sichuan Province, and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China
| | - Hui-Lin Qin
- Medical Imaging Key Laboratory of Sichuan Province, and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China
| | - Yue-Su Wang
- Medical Imaging Key Laboratory of Sichuan Province, and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China
| | - Yue-Qin Gou
- Medical Imaging Key Laboratory of Sichuan Province, and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China
| | - Hui Luo
- Medical Imaging Key Laboratory of Sichuan Province, and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China
| | - Xiao-Ming Zhang
- The First Clinical College of Jinan University, and Jinan University First Affiliated Hospital, Guangzhou, Guangdong 510630, China; Medical Imaging Key Laboratory of Sichuan Province, and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China.
| | - Tian-Wu Chen
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.
| |
Collapse
|
24
|
Elahi R, Nazari M. An updated overview of radiomics-based artificial intelligence (AI) methods in breast cancer screening and diagnosis. Radiol Phys Technol 2024; 17:795-818. [PMID: 39285146 DOI: 10.1007/s12194-024-00842-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/25/2024] [Accepted: 08/27/2024] [Indexed: 11/21/2024]
Abstract
Current imaging methods for diagnosing breast cancer (BC) are associated with limited sensitivity and specificity and modest positive predictive power. The recent progress in image analysis using artificial intelligence (AI) has created great promise to improve BC diagnosis and subtype differentiation. In this case, novel quantitative computational methods, such as radiomics, have been developed to enhance the sensitivity and specificity of early BC diagnosis and classification. The potential of radiomics in improving the diagnostic efficacy of imaging studies has been shown in several studies. In this review article, we discuss the radiomics workflow and current handcrafted radiomics methods in the diagnosis and classification of BC based on the most recent studies on different imaging modalities, e.g., MRI, mammography, contrast-enhanced spectral mammography (CESM), ultrasound imaging, and digital breast tumosynthesis (DBT). We also discuss current challenges and potential strategies to improve the specificity and sensitivity of radiomics in breast cancer to help achieve a higher level of BC classification and diagnosis in the clinical setting. The growing field of AI incorporation with imaging information has opened a great opportunity to provide a higher level of care for BC patients.
Collapse
Affiliation(s)
- Reza Elahi
- Department of Radiology, Zanjan University of Medical Sciences, Zanjan, Iran.
| | - Mahdis Nazari
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
25
|
Lee SB, Hong Y, Cho YJ, Jeong D, Lee J, Choi JW, Hwang JY, Lee S, Choi YH, Cheon JE. Enhancing Radiomics Reproducibility: Deep Learning-Based Harmonization of Abdominal Computed Tomography (CT) Images. Bioengineering (Basel) 2024; 11:1212. [PMID: 39768030 PMCID: PMC11673047 DOI: 10.3390/bioengineering11121212] [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: 10/17/2024] [Revised: 11/27/2024] [Accepted: 11/28/2024] [Indexed: 01/11/2025] Open
Abstract
We assessed the feasibility of using deep learning-based image harmonization to improve the reproducibility of radiomics features in abdominal CT scans. In CT imaging, harmonization adjusts images from different institutions to ensure consistency despite variations in scanners and acquisition protocols. This process is essential because such differences can lead to variability in radiomics features, affecting reproducibility and accuracy. Harmonizing images minimizes these inconsistencies, supporting more reliable and clinically applicable results across diverse settings. A pre-trained harmonization algorithm was applied to 63 dual-energy abdominal CT images, which were reconstructed into four different types, and 10 regions of interest (ROIs) were analyzed. From the original 455 radiomics features per ROI, 387 were used after excluding redundant features. Reproducibility was measured using the intraclass correlation coefficient (ICC), with a threshold of ICC ≥ 0.85 indicating acceptable reproducibility. The region-based analysis revealed significant improvements in reproducibility post-harmonization, especially in vessel features, which increased from 14% to 69%. Other regions, including the spleen, kidney, muscle, and liver parenchyma, also saw notable improvements, although air reproducibility slightly decreased from 95% to 94%, impacting only a few features. In patient-based analysis, reproducible features increased from 18% to 65%, with an average of 179 additional reproducible features per patient after harmonization. These results demonstrate that deep learning-based harmonization can significantly enhance the reproducibility of radiomics features in abdominal CT, offering promising potential for advancing radiomics development and its clinical applications.
Collapse
Affiliation(s)
- Seul Bi Lee
- Department of Radiology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea; (S.B.L.); (J.W.C.); (J.Y.H.); (S.L.); (Y.H.C.); (J.-E.C.)
| | - Youngtaek Hong
- CONNECT-AI R&D Center, Yonsei University College of Medicine, Seoul 03080, Republic of Korea; (Y.H.); (D.J.); (J.L.)
| | - Yeon Jin Cho
- Department of Radiology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea; (S.B.L.); (J.W.C.); (J.Y.H.); (S.L.); (Y.H.C.); (J.-E.C.)
- Department of Radiology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
| | - Dawun Jeong
- CONNECT-AI R&D Center, Yonsei University College of Medicine, Seoul 03080, Republic of Korea; (Y.H.); (D.J.); (J.L.)
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul 03080, Republic of Korea
| | - Jina Lee
- CONNECT-AI R&D Center, Yonsei University College of Medicine, Seoul 03080, Republic of Korea; (Y.H.); (D.J.); (J.L.)
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul 03080, Republic of Korea
| | - Jae Won Choi
- Department of Radiology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea; (S.B.L.); (J.W.C.); (J.Y.H.); (S.L.); (Y.H.C.); (J.-E.C.)
| | - Jae Yeon Hwang
- Department of Radiology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea; (S.B.L.); (J.W.C.); (J.Y.H.); (S.L.); (Y.H.C.); (J.-E.C.)
| | - Seunghyun Lee
- Department of Radiology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea; (S.B.L.); (J.W.C.); (J.Y.H.); (S.L.); (Y.H.C.); (J.-E.C.)
- Department of Radiology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
| | - Young Hun Choi
- Department of Radiology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea; (S.B.L.); (J.W.C.); (J.Y.H.); (S.L.); (Y.H.C.); (J.-E.C.)
- Department of Radiology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
| | - Jung-Eun Cheon
- Department of Radiology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea; (S.B.L.); (J.W.C.); (J.Y.H.); (S.L.); (Y.H.C.); (J.-E.C.)
- Department of Radiology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
- Institute of Radiation Medicine, Seoul National University Medical Research Center, 103 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
| |
Collapse
|
26
|
Shenouda M, Shaikh A, Deutsch I, Mitchell O, Kindler HL, Armato SG. Radiomics for differentiation of somatic BAP1 mutation on CT scans of patients with pleural mesothelioma. J Med Imaging (Bellingham) 2024; 11:064501. [PMID: 39669009 PMCID: PMC11633667 DOI: 10.1117/1.jmi.11.6.064501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 11/21/2024] [Accepted: 11/21/2024] [Indexed: 12/14/2024] Open
Abstract
Purpose The BRCA1-associated protein 1 (BAP1) gene is of great interest because somatic (BAP1) mutations are the most common alteration associated with pleural mesothelioma (PM). Further, germline mutation of the BAP1 gene has been linked to the development of PM. This study aimed to explore the potential of radiomics on computed tomography scans to identify somatic BAP1 gene mutations and assess the feasibility of radiomics in future research in identifying germline mutations. Approach A cohort of 149 patients with PM and known somatic BAP1 mutation status was collected, and a previously published deep learning model was used to first automatically segment the tumor, followed by radiologist modifications. Image preprocessing was performed, and texture features were extracted from the segmented tumor regions. The top features were selected and used to train 18 separate machine learning models using leave-one-out cross-validation (LOOCV). The performance of the models in distinguishing between BAP1-mutated (BAP1+) and BAP1 wild-type (BAP1-) tumors was evaluated using the receiver operating characteristic area under the curve (ROC AUC). Results A decision tree classifier achieved the highest overall AUC value of 0.69 (95% confidence interval: 0.60 and 0.77). The features selected most frequently through the LOOCV were all second-order (gray-level co-occurrence or gray-level size zone matrices) and were extracted from images with an applied transformation. Conclusions This proof-of-concept work demonstrated the potential of radiomics to differentiate among BAP1+/- in patients with PM. Future work will extend these methods to the assessment of germline BAP1 mutation status through image analysis for improved patient prognostication.
Collapse
Affiliation(s)
- Mena Shenouda
- The University of Chicago, Department of Radiology, Chicago, Illinois, United States
| | | | - Ilana Deutsch
- Northwestern University, Evanston, Illinois, United States
| | - Owen Mitchell
- The University of Chicago, Department of Medicine, Chicago, Illinois, United States
| | - Hedy L. Kindler
- The University of Chicago, Department of Medicine, Chicago, Illinois, United States
| | - Samuel G. Armato
- The University of Chicago, Department of Radiology, Chicago, Illinois, United States
| |
Collapse
|
27
|
Mayerhoefer ME, Shepherd TM, Weber M, Leithner D, Woo S, Pan JW, Pardoe HR. Sexual Dimorphism of Radiomic Features in the Brain: An Exploratory Study Using 700 μm MP2RAGE MRI at 7 T. Invest Radiol 2024; 59:782-786. [PMID: 38896439 DOI: 10.1097/rli.0000000000001088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
OBJECTIVES The aim of this study was to determine whether MRI radiomic features of key cerebral structures differ between women and men, and whether detection of such differences depends on the image resolution. MATERIALS AND METHODS Ultrahigh resolution (UHR) 3D MP2RAGE (magnetization-prepared 2 rapid acquisition gradient echo) T1-weighted MR images (voxel size, 0.7 × 0.7 × 0.7 mm 3 ) of the brain of 30 subjects (18 women and 12 men; mean age, 39.0 ± 14.8 years) without abnormal findings on MRI were retrospectively included. MRI was performed on a whole-body 7 T MR system. A convolutional neural network was used to segment the following structures: frontal cortex, frontal white matter, thalamus, putamen, globus pallidus, caudate nucleus, and corpus callosum. Eighty-seven radiomic features were extracted respectively: gray-level histogram (n = 18), co-occurrence matrix (n = 24), run-length matrix (n = 16), size-zone matrix (n = 16), and dependence matrix (n = 13). Feature extraction was performed at UHR and, additionally, also after resampling to 1.4 × 1.4 × 1.4 mm 3 voxel size (standard clinical resolution). Principal components (PCs) of radiomic features were calculated, and independent samples t tests with Cohen d as effect size measure were used to assess differences in PCs between women and men for the different cerebral structures. RESULTS At UHR, at least a single PC differed significantly between women and men in 6/7 cerebral structures: frontal cortex ( d = -0.79, P = 0.042 and d = -1.01, P = 0.010), frontal white matter ( d = -0.81, P = 0.039), thalamus ( d = 1.43, P < 0.001), globus pallidus ( d = 0.92, P = 0.020), caudate nucleus ( d = -0.83, P = 0.039), and corpus callosum ( d = -0.97, P = 0.039). At standard clinical resolution, only a single PC extracted from the corpus callosum differed between sexes ( d = 1.05, P = 0.009). CONCLUSIONS Nonnegligible differences in radiomic features of several key structures of the brain exist between women and men, and need to be accounted for. Very high spatial resolution may be required to uncover and further investigate the sexual dimorphism of brain structures on MRI.
Collapse
Affiliation(s)
- Marius E Mayerhoefer
- From the Department of Radiology, NYU Grossman School of Medicine, New York, NY (M.E.M., T.M.S., D.L., S.W.); Division of General and Pediatric Radiology, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria (M.E.M., M.W.); Florey Institute of Neuroscience and Mental Health, Melbourne, Australia (H.R.P.); Comprehensive Epilepsy Center, Department of Neurology, NYU Grossman School of Medicine, New York, NY (H.R.P.); and Department of Radiology, University of Missouri Columbia, Columbia, MO (J.W.P.)
| | | | | | | | | | | | | |
Collapse
|
28
|
Jung HK, Kim K, Park JE, Kim N. Image-Based Generative Artificial Intelligence in Radiology: Comprehensive Updates. Korean J Radiol 2024; 25:959-981. [PMID: 39473088 PMCID: PMC11524689 DOI: 10.3348/kjr.2024.0392] [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: 04/19/2024] [Revised: 08/29/2024] [Accepted: 08/29/2024] [Indexed: 11/02/2024] Open
Abstract
Generative artificial intelligence (AI) has been applied to images for image quality enhancement, domain transfer, and augmentation of training data for AI modeling in various medical fields. Image-generative AI can produce large amounts of unannotated imaging data, which facilitates multiple downstream deep-learning tasks. However, their evaluation methods and clinical utility have not been thoroughly reviewed. This article summarizes commonly used generative adversarial networks and diffusion models. In addition, it summarizes their utility in clinical tasks in the field of radiology, such as direct image utilization, lesion detection, segmentation, and diagnosis. This article aims to guide readers regarding radiology practice and research using image-generative AI by 1) reviewing basic theories of image-generative AI, 2) discussing the methods used to evaluate the generated images, 3) outlining the clinical and research utility of generated images, and 4) discussing the issue of hallucinations.
Collapse
Affiliation(s)
- Ha Kyung Jung
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Kiduk Kim
- Department of Convergence Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Ji Eun Park
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea.
| | - Namkug Kim
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
- Department of Convergence Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea.
| |
Collapse
|
29
|
Mariani I, Maino C, Giandola TP, Franco PN, Drago SG, Corso R, Talei Franzesi C, Ippolito D. Texture Analysis and Prediction of Response to Neoadjuvant Treatment in Patients with Locally Advanced Rectal Cancer. GASTROINTESTINAL DISORDERS 2024; 6:858-870. [DOI: 10.3390/gidisord6040060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2025] Open
Abstract
Background: The purpose of this study is to determine the relationship between the texture analysis extracted from preoperative rectal magnetic resonance (MR) studies and the response to neoadjuvant treatment. Materials and Methods: In total, 88 patients with rectal adenocarcinoma who underwent staging MR between 2017 and 2022 were retrospectively enrolled. After the completion of neoadjuvant treatment, they underwent surgical resection. The tumour regression grade (TRG) was collected. Patients with TRG 1–2 were classified as responders, while patients with TRG 3 to 5 were classified as non-responders. A texture analysis was conducted using LIFEx software (v 7.6.0), where T2-weighted MR sequences on oriented axial planes were uploaded, and a region of interest (ROI) was manually drawn on a single slice. Features with a Spearman correlation index > 0.5 have been discarded, and a LASSO feature selection has been applied. Selected features were trained using bootstrapping. Results: According to the TRG classes, 49 patients (55.8%) were considered responders, while 39 (44.2) were non-responders. Two features were associated with the responder class: GLCM_Homogeneity and Discretized Histo Entropy log 2. Regarding GLCM_Homogeneity, the area under the receiver operating characteristic curve (AUC), sensitivity, and specificity were 0.779 (95% CIs = 0.771–0.816), 86% (80–90), and 67% (60–71). Regarding Discretized Histo Entropy log 2, we found 0.775 AUC (0.700–0.801), 80% sensitivity (74–83), and 63% specificity (58–69). Combining both radiomics features the radiomics signature diagnostic accuracy increased (AUC = 0.844). Finally, the AUC of 1000 bootstraps were 0.810. Conclusions: Texture analysis can be considered an advanced tool for determining a possible correlation between pre-surgical MR data and the response to neoadjuvant therapy.
Collapse
Affiliation(s)
- Ilaria Mariani
- Department of Diagnostic Radiology, IRCCS Fondazione San Gerardo dei Tintori, Via Pergolesi 33, 20900 Monza, Italy
| | - Cesare Maino
- Department of Diagnostic Radiology, IRCCS Fondazione San Gerardo dei Tintori, Via Pergolesi 33, 20900 Monza, Italy
| | - Teresa Paola Giandola
- Department of Diagnostic Radiology, IRCCS Fondazione San Gerardo dei Tintori, Via Pergolesi 33, 20900 Monza, Italy
| | - Paolo Niccolò Franco
- Department of Diagnostic Radiology, IRCCS Fondazione San Gerardo dei Tintori, Via Pergolesi 33, 20900 Monza, Italy
| | - Silvia Girolama Drago
- Department of Diagnostic Radiology, IRCCS Fondazione San Gerardo dei Tintori, Via Pergolesi 33, 20900 Monza, Italy
| | - Rocco Corso
- Department of Diagnostic Radiology, IRCCS Fondazione San Gerardo dei Tintori, Via Pergolesi 33, 20900 Monza, Italy
| | - Cammillo Talei Franzesi
- Department of Diagnostic Radiology, IRCCS Fondazione San Gerardo dei Tintori, Via Pergolesi 33, 20900 Monza, Italy
| | - Davide Ippolito
- Department of Diagnostic Radiology, IRCCS Fondazione San Gerardo dei Tintori, Via Pergolesi 33, 20900 Monza, Italy
- School of Medicine, University of Milano Bicocca, Via Cadore 33, 20090 Monza, Italy
| |
Collapse
|
30
|
Mahmoud M, Lin KH, Lee RC, Liu CA. Assessment of Y-90 Radioembolization Treatment Response for Hepatocellular Carcinoma Cases Using MRI Radiomics. Mol Imaging Radionucl Ther 2024; 33:156-166. [PMID: 39373149 PMCID: PMC11589346 DOI: 10.4274/mirt.galenos.2024.59365] [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: 10/19/2023] [Accepted: 08/04/2024] [Indexed: 10/08/2024] Open
Abstract
Objectives This study aimed to investigate the ability of radiomics features extracted from magnetic resonance imaging (MRI) images to differentiate between responders and non-responders for hepatocellular carcinoma (HCC) cases who received Y-90 transarterial radioembolization treatment. Methods Thirty-six cases of HCC who underwent MRI scans after Y-90 radioembolization were included in this study. Tumors were segmented from MRI T2 images, and then 87 radiomic features were extracted through the LIFEx package software. Treatment response was determined 9 months after treatment through the modified response evaluation criteria in solid tumours (mRECIST). Results According to mRECIST, 28 cases were responders and 8 cases were non-responders. Two radiomics features, "Grey Level Size Zone Matrix (GLSZM)-Small Zone Emphasis" and "GLSZM-Normalized Zone Size Non-Uniformity", were the radiomics features that could predict treatment response with the area under curve (AUC)= 0.71, sensitivity= 0.93, and specificity= 0.62 for both features. Whereas the other 4 features (kurtosis, intensity histogram root mean square, neighbourhood gray-tone difference matrix strength, and GLSZM normalized grey level non-uniformity) have a relatively lower but acceptable discrimination ability range from AUC= 0.6 to 0.66. Conclusion MRI radiomics analysis could be used to assess the treatment response for HCC cases treated with Y-90 radioembolization.
Collapse
Affiliation(s)
| | - Ko-Han Lin
- Taipei Veterans General Hospital, Clinic of Nuclear Medicine, Taipei, Taiwan
| | - Rheun-Chuan Lee
- Taipei Veterans General Hospital, Clinic of Radiology, Taipei, Taiwan
| | - Chien-an Liu
- Taipei Veterans General Hospital, Clinic of Radiology, Taipei, Taiwan
| |
Collapse
|
31
|
Soleymani Y, Valibeiglou Z, Fazel Ghaziani M, Jahanshahi A, Khezerloo D. Radiomics reproducibility in computed tomography through changes of ROI size, resolution, and hounsfield unit: A phantom study. Radiography (Lond) 2024; 30:1629-1636. [PMID: 39423630 DOI: 10.1016/j.radi.2024.10.003] [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] [Revised: 09/23/2024] [Accepted: 10/02/2024] [Indexed: 10/21/2024]
Abstract
INTRODUCTION Although radiomics has revealed an intriguing perspective for quantitative radiology, the impact of scanning parameters on its outcomes must be considered. In this study, the effects of changes in the region of interest (ROI) sizes, Hounsfield Unit (HU), and resolution of computed tomography (CT) on feature reproducibility have been investigated. METHODS The GAMMEX 464 phantom was used to evaluate the reproducibility of radiomics features across different ROI sizes, HU, and resolution. Data were acquired using a consistent system setup, with the phantom repositioned for each scan. The first acquisition series examined the effects of different ROI sizes and resolutions (1, 3, and 5 mm) on feature reproducibility. The second series assessed the impact of different HU and resolution. Segmentation and feature extraction were performed using LIFEx 7.1.0 software, focusing on textural radiomics features. Statistical analysis involved calculating the coefficient of variation (COV) to categorize feature variability. COV <5 % was considered highly stable. RESULTS Out of the 32 textural features studied, the analysis of changes in ROI size with a resolution of 1 mm, 3 mm, and 5 mm revealed that 16, 17, and 18 features had high reproducibility, with a COV<5 %. Polyethylene, acrylic, and water also demonstrated stable textural features across changes in scan parameters and image resolutions, with 4 reproducible features in all resolutions. The grey-level run length matrix (GLRLM) and grey-level zone length matrix (GLZLM) radiomics groups were highly stable in the context of variations in scan parameters and different materials. CONCLUSION The results of this study highlight the importance of standardizing radiomics studies to reduce the influence of pre-analysis steps on feature values. This standardization is crucial for guaranteeing the consistency of radiomics features under various imaging conditions. Additional research is required to enhance these results. IMPLICATIONS FOR PRACTICE To ensure the reproducibility and reliability of radiomics features, it is imperative to standardize scanning parameters and pre-analysis protocols. This standardization will enhance the consistency of radiomics applications in both clinical and research environments.
Collapse
Affiliation(s)
- Y Soleymani
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Z Valibeiglou
- Department of Medical Physics, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - M Fazel Ghaziani
- Department of Radiology, Faculty of Allied Medical Sciences, Tabriz University of Medical Science, Tabriz, Iran
| | - A Jahanshahi
- Department of Radiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - D Khezerloo
- Department of Radiology, Faculty of Allied Medical Sciences, Tabriz University of Medical Science, Tabriz, Iran.
| |
Collapse
|
32
|
Coppes RP, van Dijk LV. Future of Team-based Basic and Translational Science in Radiation Oncology. Semin Radiat Oncol 2024; 34:370-378. [PMID: 39271272 DOI: 10.1016/j.semradonc.2024.07.007] [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: 09/15/2024]
Abstract
To further optimize radiotherapy, a more personalized treatment towards individual patient's risk profiles, dissecting both patient-specific tumor and normal tissue response to multimodality treatments is needed. Novel developments in radiobiology, using in vitro patient-specific complex tissue resembling 3D models and multiomics approaches at a spatial single-cell level, may provide unprecedented insight into the radiation responses of tumors and normal tissue. Here, we describe the necessary team effort, including all disciplines in radiation oncology, to integrate such data into clinical prediction models and link the relatively "big data" from the clinical practice, allowing accurate patient stratification for personalized treatment approaches.
Collapse
Affiliation(s)
- R P Coppes
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.; Department of Biomedical Sciences, University Medical Center Groningen, University of Groningen, Groningen, Netherlands..
| | - L V van Dijk
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
33
|
Zhu N, Meng X, Wang Z, Hu Y, Zhao T, Fan H, Niu F, Han J. Radiomics in Diagnosis, Grading, and Treatment Response Assessment of Soft Tissue Sarcomas: A Systematic Review and Meta-analysis. Acad Radiol 2024; 31:3982-3992. [PMID: 38772802 DOI: 10.1016/j.acra.2024.03.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 03/12/2024] [Accepted: 03/22/2024] [Indexed: 05/23/2024]
Abstract
RATIONALE AND OBJECTIVES To evaluate radiomics in soft tissue sarcomas (STSs) for diagnostic accuracy, grading, and treatment response assessment, with a focus on clinical relevance. METHODS In this diagnostic accuracy study, radiomics was applied using multiple MRI sequences and AI classifiers, with histopathological diagnosis as the reference standard. Statistical analysis involved meta-analysis, random-effects model, and Deeks' funnel plot asymmetry test. RESULTS Among 579 unique titles and abstracts, 24 articles were included in the systematic review, with 21 used for meta-analysis. Radiomics demonstrated a pooled sensitivity of 84% (95% CI: 80-87) and specificity of 63% (95% CI: 56-70), AUC of 0.93 for diagnosis, sensitivity of 84% (95% CI: 82-87) and specificity of 73% (95% CI: 68-77), AUC of 0.91 for grading, and sensitivity of 83% (95% CI: 67-94) and specificity of 67% (95% CI: 59-74), AUC of 0.87 for treatment response assessment. CONCLUSION Radiomics exhibits potential for accurate diagnosis, grading, and treatment response assessment in STSs, emphasizing the need for standardization and prospective trials. CLINICAL RELEVANCE STATEMENT Radiomics offers precise tools for STS diagnosis, grading, and treatment response assessment, with implications for optimizing patient care and treatment strategies in this complex malignancy.
Collapse
Affiliation(s)
- Nana Zhu
- The Department of Radiology, Tianjin Hospital, 406 Jiefang Southern Road, Tianjin, China; Graduate School, Tianjin Medical University, Tianjin, China
| | - Xianghong Meng
- The Department of Radiology, Tianjin Hospital, 406 Jiefang Southern Road, Tianjin, China
| | - Zhi Wang
- The Department of Radiology, Tianjin Hospital, 406 Jiefang Southern Road, Tianjin, China; Graduate School, Tianjin Medical University, Tianjin, China.
| | - Yongcheng Hu
- The Department of Radiology, Tianjin Hospital, 406 Jiefang Southern Road, Tianjin, China; Graduate School, Tianjin Medical University, Tianjin, China
| | - Tingting Zhao
- The Department of Radiology, Tianjin Hospital, 406 Jiefang Southern Road, Tianjin, China; Graduate School, Tianjin Medical University, Tianjin, China
| | - Hongxing Fan
- The Department of Radiology, Tianjin Hospital, 406 Jiefang Southern Road, Tianjin, China; Graduate School, Tianjin Medical University, Tianjin, China
| | - Feige Niu
- The Department of Radiology, Tianjin Hospital, 406 Jiefang Southern Road, Tianjin, China; Graduate School, Tianjin Medical University, Tianjin, China
| | - Jun Han
- The Department of Radiology, Tianjin Hospital, 406 Jiefang Southern Road, Tianjin, China; Graduate School, Tianjin University, Tianjin, China
| |
Collapse
|
34
|
Tran K, Ginzburg D, Hong W, Attenberger U, Ko HS. Post-radiotherapy stage III/IV non-small cell lung cancer radiomics research: a systematic review and comparison of CLEAR and RQS frameworks. Eur Radiol 2024; 34:6527-6543. [PMID: 38625613 PMCID: PMC11399214 DOI: 10.1007/s00330-024-10736-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 02/07/2024] [Accepted: 03/04/2024] [Indexed: 04/17/2024]
Abstract
BACKGROUND Lung cancer, the second most common cancer, presents persistently dismal prognoses. Radiomics, a promising field, aims to provide novel imaging biomarkers to improve outcomes. However, clinical translation faces reproducibility challenges, despite efforts to address them with quality scoring tools. OBJECTIVE This study had two objectives: 1) identify radiomics biomarkers in post-radiotherapy stage III/IV nonsmall cell lung cancer (NSCLC) patients, 2) evaluate research quality using the CLEAR (CheckList_for_EvaluAtion_of_Radiomics_research), RQS (Radiomics_Quality_Score) frameworks, and formulate an amalgamated CLEAR-RQS tool to enhance scientific rigor. MATERIALS AND METHODS A systematic literature review (Jun-Aug 2023, MEDLINE/PubMed/SCOPUS) was conducted concerning stage III/IV NSCLC, radiotherapy, and radiomic features (RF). Extracted data included study design particulars, such as sample size, radiotherapy/CT technique, selected RFs, and endpoints. CLEAR and RQS were merged into a CLEAR-RQS checklist. Three readers appraised articles utilizing CLEAR, RQS, and CLEAR-RQS metrics. RESULTS Out of 871 articles, 11 met the inclusion/exclusion criteria. The Median cohort size was 91 (range: 10-337) with 9 studies being single-center. No common RF were identified. The merged CLEAR-RQS checklist comprised 61 items. Most unreported items were within CLEAR's "methods" and "open-source," and within RQS's "phantom-calibration," "registry-enrolled prospective-trial-design," and "cost-effective-analysis" sections. No study scored above 50% on RQS. Median CLEAR scores were 55.74% (32.33/58 points), and for RQS, 17.59% (6.3/36 points). CLEAR-RQS article ranking fell between CLEAR and RQS and aligned with CLEAR. CONCLUSION Radiomics research in post-radiotherapy stage III/IV NSCLC exhibits variability and frequently low-quality reporting. The formulated CLEAR-RQS checklist may facilitate education and holds promise for enhancing radiomics research quality. CLINICAL RELEVANCE STATEMENT Current radiomics research in the field of stage III/IV postradiotherapy NSCLC is heterogenous, lacking reproducibility, with no identified imaging biomarker. Radiomics research quality assessment tools may enhance scientific rigor and thereby facilitate radiomics translation into clinical practice. KEY POINTS There is heterogenous and low radiomics research quality in postradiotherapy stage III/IV nonsmall cell lung cancer. Barriers to reproducibility are small cohort size, nonvalidated studies, missing technical parameters, and lack of data, code, and model sharing. CLEAR (CheckList_for_EvaluAtion_of_Radiomics_research), RQS (Radiomics_Quality_Score), and the amalgamated CLEAR-RQS tool are useful frameworks for assessing radiomics research quality and may provide a valuable resource for educational purposes in the field of radiomics.
Collapse
Affiliation(s)
- Kevin Tran
- Department of Cancer Imaging, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
- Faculty of Medicine, Dentistry & Health Sciences, University of Melbourne, Parkville, VIC 3052, Australia
| | - Daniel Ginzburg
- Department of Diagnostic and Interventional Radiology, Venusberg Campus 1, 53127, Bonn, Germany
| | - Wei Hong
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - Ulrike Attenberger
- Department of Diagnostic and Interventional Radiology, Venusberg Campus 1, 53127, Bonn, Germany
| | - Hyun Soo Ko
- Department of Cancer Imaging, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia.
- Department of Diagnostic and Interventional Radiology, Venusberg Campus 1, 53127, Bonn, Germany.
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, 305 Grattan St, Melbourne, VIC 3000, Australia.
| |
Collapse
|
35
|
Badesha AS, Frood R, Bailey MA, Coughlin PM, Scarsbrook AF. A Scoping Review of Machine-Learning Derived Radiomic Analysis of CT and PET Imaging to Investigate Atherosclerotic Cardiovascular Disease. Tomography 2024; 10:1455-1487. [PMID: 39330754 PMCID: PMC11435603 DOI: 10.3390/tomography10090108] [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: 07/25/2024] [Revised: 08/27/2024] [Accepted: 08/30/2024] [Indexed: 09/28/2024] Open
Abstract
BACKGROUND Cardiovascular disease affects the carotid arteries, coronary arteries, aorta and the peripheral arteries. Radiomics involves the extraction of quantitative data from imaging features that are imperceptible to the eye. Radiomics analysis in cardiovascular disease has largely focused on CT and MRI modalities. This scoping review aims to summarise the existing literature on radiomic analysis techniques in cardiovascular disease. METHODS MEDLINE and Embase databases were searched for eligible studies evaluating radiomic techniques in living human subjects derived from CT, MRI or PET imaging investigating atherosclerotic disease. Data on study population, imaging characteristics and radiomics methodology were extracted. RESULTS Twenty-nine studies consisting of 5753 patients (3752 males) were identified, and 78.7% of patients were from coronary artery studies. Twenty-seven studies employed CT imaging (19 CT carotid angiography and 6 CT coronary angiography (CTCA)), and two studies studied PET/CT. Manual segmentation was most frequently undertaken. Processing techniques included voxel discretisation, voxel resampling and filtration. Various shape, first-order, second-order and higher-order radiomic features were extracted. Logistic regression was most commonly used for machine learning. CONCLUSION Most published evidence was feasibility/proof of concept work. There was significant heterogeneity in image acquisition, segmentation techniques, processing and analysis between studies. There is a need for the implementation of standardised imaging acquisition protocols, adherence to published reporting guidelines and economic evaluation.
Collapse
Affiliation(s)
- Arshpreet Singh Badesha
- Department of Radiology, St. James’s University Hospital, Leeds Teaching Hospitals NHS Trust, Leeds LS9 7TF, UK
| | - Russell Frood
- Department of Radiology, St. James’s University Hospital, Leeds Teaching Hospitals NHS Trust, Leeds LS9 7TF, UK
- Faculty of Medicine and Health, University of Leeds, Leeds LS2 9TJ, UK
| | - Marc A. Bailey
- Faculty of Medicine and Health, University of Leeds, Leeds LS2 9TJ, UK
- The Leeds Vascular Institute, Leeds General Infirmary, Leeds Teaching Hospitals NHS Trust, Leeds LS1 3EX, UK
| | - Patrick M. Coughlin
- The Leeds Vascular Institute, Leeds General Infirmary, Leeds Teaching Hospitals NHS Trust, Leeds LS1 3EX, UK
| | - Andrew F. Scarsbrook
- Department of Radiology, St. James’s University Hospital, Leeds Teaching Hospitals NHS Trust, Leeds LS9 7TF, UK
- Faculty of Medicine and Health, University of Leeds, Leeds LS2 9TJ, UK
| |
Collapse
|
36
|
Fajemisin JA, Gonzalez G, Rosenberg SA, Ullah G, Redler G, Latifi K, Moros EG, El Naqa I. Magnetic Resonance-Guided Cancer Therapy Radiomics and Machine Learning Models for Response Prediction. Tomography 2024; 10:1439-1454. [PMID: 39330753 PMCID: PMC11435563 DOI: 10.3390/tomography10090107] [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: 08/08/2024] [Revised: 08/27/2024] [Accepted: 08/28/2024] [Indexed: 09/28/2024] Open
Abstract
Magnetic resonance imaging (MRI) is known for its accurate soft tissue delineation of tumors and normal tissues. This development has significantly impacted the imaging and treatment of cancers. Radiomics is the process of extracting high-dimensional features from medical images. Several studies have shown that these extracted features may be used to build machine-learning models for the prediction of treatment outcomes of cancer patients. Various feature selection techniques and machine models interrogate the relevant radiomics features for predicting cancer treatment outcomes. This study aims to provide an overview of MRI radiomics features used in predicting clinical treatment outcomes with machine learning techniques. The review includes examples from different disease sites. It will also discuss the impact of magnetic field strength, sample size, and other characteristics on outcome prediction performance.
Collapse
Affiliation(s)
- Jesutofunmi Ayo Fajemisin
- Department of Physics, University of South Florida, Tampa, FL 33620, USA
- Machine Learning Department, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Glebys Gonzalez
- Machine Learning Department, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Stephen A Rosenberg
- Machine Learning Department, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
- Radiation Oncology Department, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Ghanim Ullah
- Department of Physics, University of South Florida, Tampa, FL 33620, USA
| | - Gage Redler
- Radiation Oncology Department, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Kujtim Latifi
- Radiation Oncology Department, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Eduardo G Moros
- Department of Physics, University of South Florida, Tampa, FL 33620, USA
- Machine Learning Department, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
- Radiation Oncology Department, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Issam El Naqa
- Department of Physics, University of South Florida, Tampa, FL 33620, USA
- Machine Learning Department, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| |
Collapse
|
37
|
Wu Y, Cao F, Lei H, Zhang S, Mei H, Ni L, Pang J. Interpretable multiphasic CT-based radiomic analysis for preoperatively differentiating benign and malignant solid renal tumors: a multicenter study. Abdom Radiol (NY) 2024; 49:3096-3106. [PMID: 38733392 PMCID: PMC11335970 DOI: 10.1007/s00261-024-04351-3] [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: 03/08/2024] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 05/13/2024]
Abstract
BACKGROUND To develop and compare machine learning models based on triphasic contrast-enhanced CT (CECT) for distinguishing between benign and malignant renal tumors. MATERIALS AND METHODS In total, 427 patients were enrolled from two medical centers: Center 1 (serving as the training set) and Center 2 (serving as the external validation set). First, 1781 radiomic features were individually extracted from corticomedullary phase (CP), nephrographic phase (NP), and excretory phase (EP) CECT images, after which 10 features were selected by the minimum redundancy maximum relevance method. Second, random forest (RF) models were constructed from single-phase features (CP, NP, and EP) as well as from the combination of features from all three phases (TP). Third, the RF models were assessed in the training and external validation sets. Finally, the internal prediction mechanisms of the models were explained by the SHapley Additive exPlanations (SHAP) approach. RESULTS A total of 266 patients with renal tumors from Center 1 and 161 patients from Center 2 were included. In the training set, the AUCs of the RF models constructed from the CP, NP, EP, and TP features were 0.886, 0.912, 0.930, and 0.944, respectively. In the external validation set, the models achieved AUCs of 0.860, 0.821, 0.921, and 0.908, respectively. The "original_shape_Flatness" feature played the most important role in the prediction outcome for the RF model based on EP features according to the SHAP method. CONCLUSIONS The four RF models efficiently differentiated benign from malignant solid renal tumors, with the EP feature-based RF model displaying the best performance.
Collapse
Affiliation(s)
- Yaohai Wu
- Department of Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Fei Cao
- Department of Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Hanqi Lei
- Department of Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Shiqiang Zhang
- Department of Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Hongbing Mei
- Department of Urology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Liangchao Ni
- Department of Urology, Guangdong and Shenzhen Key Laboratory of Reproductive Medicine and Genetics, Peking University Shenzhen Hospital, Shenzhen, China
| | - Jun Pang
- Department of Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China.
| |
Collapse
|
38
|
Chen SQ, Wei L, He K, Xiao YW, Zhang ZT, Dai JK, Shu T, Sun XY, Wu D, Luo Y, Gui YF, Xiao XL. A radiomics nomogram based on multiparametric MRI for diagnosing focal cortical dysplasia and initially identifying laterality. BMC Med Imaging 2024; 24:216. [PMID: 39148028 PMCID: PMC11325615 DOI: 10.1186/s12880-024-01374-6] [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: 11/26/2022] [Accepted: 07/22/2024] [Indexed: 08/17/2024] Open
Abstract
BACKGROUND Focal cortical dysplasia (FCD) is the most common epileptogenic developmental malformation. The diagnosis of FCD is challenging. We generated a radiomics nomogram based on multiparametric magnetic resonance imaging (MRI) to diagnose FCD and identify laterality early. METHODS Forty-three patients treated between July 2017 and May 2022 with histopathologically confirmed FCD were retrospectively enrolled. The contralateral unaffected hemispheres were included as the control group. Therefore, 86 ROIs were finally included. Using January 2021 as the time cutoff, those admitted after January 2021 were included in the hold-out set (n = 20). The remaining patients were separated randomly (8:2 ratio) into training (n = 55) and validation (n = 11) sets. All preoperative and postoperative MR images, including T1-weighted (T1w), T2-weighted (T2w), fluid-attenuated inversion recovery (FLAIR), and combined (T1w + T2w + FLAIR) images, were included. The least absolute shrinkage and selection operator (LASSO) was used to select features. Multivariable logistic regression analysis was used to develop the diagnosis model. The performance of the radiomic nomogram was evaluated with an area under the curve (AUC), net reclassification improvement (NRI), integrated discrimination improvement (IDI), calibration and clinical utility. RESULTS The model-based radiomics features that were selected from combined sequences (T1w + T2w + FLAIR) had the highest performances in all models and showed better diagnostic performance than inexperienced radiologists in the training (AUCs: 0.847 VS. 0.664, p = 0.008), validation (AUC: 0.857 VS. 0.521, p = 0.155), and hold-out sets (AUCs: 0.828 VS. 0.571, p = 0.080). The positive values of NRI (0.402, 0.607, 0.424) and IDI (0.158, 0.264, 0.264) in the three sets indicated that the diagnostic performance of Model-Combined improved significantly. The radiomics nomogram fit well in calibration curves (p > 0.05), and decision curve analysis further confirmed the clinical usefulness of the nomogram. Additionally, the contrast (the radiomics feature) of the FCD lesions not only played a crucial role in the classifier but also had a significant correlation (r = -0.319, p < 0.05) with the duration of FCD. CONCLUSION The radiomics nomogram generated by logistic regression model-based multiparametric MRI represents an important advancement in FCD diagnosis and treatment.
Collapse
Affiliation(s)
- Shi-Qi Chen
- Department of Radiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Liang Wei
- Department of Pediatrics, The Affiliated Hospital of Jinggangshan University, Jinggangshan, Jiangxi Province, China
| | - Keng He
- Department of Radiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Ya-Wen Xiao
- Department of Radiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Zhao-Tao Zhang
- Department of Radiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Jian-Kun Dai
- GE Healthcare, MR Research China, Beijing, China
| | - Ting Shu
- Department of Radiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Xiao-Yu Sun
- Department of Radiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Di Wu
- Department of Radiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Yi Luo
- Department of Radiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Yi-Fei Gui
- Department of Radiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Xin-Lan Xiao
- Department of Radiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China.
| |
Collapse
|
39
|
Li B, Zhu J, Wang Y, Xu Y, Gao Z, Shi H, Nie P, Zhang J, Zhuang Y, Wang Z, Yang G. Radiomics nomogram based on CT radiomics features and clinical factors for prediction of Ki-67 expression and prognosis in clear cell renal cell carcinoma: a two-center study. Cancer Imaging 2024; 24:103. [PMID: 39107799 PMCID: PMC11302839 DOI: 10.1186/s40644-024-00744-1] [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: 01/08/2024] [Accepted: 07/24/2024] [Indexed: 08/10/2024] Open
Abstract
OBJECTIVES To develop and validate a radiomics nomogram combining radiomics features and clinical factors for preoperative evaluation of Ki-67 expression status and prognostic prediction in clear cell renal cell carcinoma (ccRCC). METHODS Two medical centers of 185 ccRCC patients were included, and each of them formed a training group (n = 130) and a validation group (n = 55). The independent predictor of Ki-67 expression status was identified by univariate and multivariate regression, and radiomics features were extracted from the preoperative CT images. The maximum relevance minimum redundancy (mRMR) and the least absolute shrinkage and selection operator algorithm (LASSO) were used to identify the radiomics features that were most relevant for high Ki-67 expression. Subsequently, clinical model, radiomics signature (RS), and radiomics nomogram were established. The performance for prediction of Ki-67 expression status was validated using area under curve (AUC), calibration curve, Delong test, decision curve analysis (DCA). Prognostic prediction was assessed by survival curve and concordance index (C-index). RESULTS Tumour size was the only independent predictor of Ki-67 expression status. Five radiomics features were finally identified to construct the RS (AUC: training group, 0.821; validation group, 0.799). The radiomics nomogram achieved a higher AUC (training group, 0.841; validation group, 0.814) and clinical net benefit. Besides, the radiomics nomogram provided a highest C-index (training group, 0.841; validation group, 0.820) in predicting prognosis for ccRCC patients. CONCLUSIONS The radiomics nomogram can accurately predict the Ki-67 expression status and exhibit a great capacity for prognostic prediction in patients with ccRCC and may provide value for tailoring personalized treatment strategies and facilitating comprehensive clinical monitoring for ccRCC patients.
Collapse
Affiliation(s)
- Ben Li
- Department of Nuclear Medicine, The Affiliated Hospital of Qingdao University, No. 59, Haier Road, Qingdao, 266061, Shandong, China
- School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Jie Zhu
- Department of Scientific Research Management and Foreign Affairs, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yanmei Wang
- GE Healthcare China, Pudong New Town, Shanghai, China
| | - Yuchao Xu
- School of Nuclear Science and Technology, University of South China, Hengyang, Hunan, China
| | - Zhaisong Gao
- Department of Nuclear Medicine, The Affiliated Hospital of Qingdao University, No. 59, Haier Road, Qingdao, 266061, Shandong, China
| | - Hailei Shi
- Department of Pathology, The Affiliated Hospital of Qingdao University, No. 16, Jiangsu Road, Qingdao, 266003, Shandong, China
| | - Pei Nie
- Department of Radiology, The Affiliated Hospital of Qingdao University, No. 16, Jiangsu Road, Qingdao, 266003, Shandong, China
| | - Ju Zhang
- Department of Nuclear Medicine, The Affiliated Hospital of Qingdao University, No. 59, Haier Road, Qingdao, 266061, Shandong, China
| | - Yuan Zhuang
- Department of Nuclear Medicine, The Affiliated Hospital of Qingdao University, No. 59, Haier Road, Qingdao, 266061, Shandong, China
| | - Zhenguang Wang
- Department of Nuclear Medicine, The Affiliated Hospital of Qingdao University, No. 59, Haier Road, Qingdao, 266061, Shandong, China.
| | - Guangjie Yang
- Department of Nuclear Medicine, The Affiliated Hospital of Qingdao University, No. 59, Haier Road, Qingdao, 266061, Shandong, China.
| |
Collapse
|
40
|
Yang F, Feng Y, Sun P, Traverso A, Dekker A, Zhang B, Huang Z, Wang Z, Yan D. Preoperative prediction of high-grade osteosarcoma response to neoadjuvant therapy based on a plain CT radiomics model: A dual-center study. J Bone Oncol 2024; 47:100614. [PMID: 38975332 PMCID: PMC11225658 DOI: 10.1016/j.jbo.2024.100614] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 05/05/2024] [Accepted: 06/01/2024] [Indexed: 07/09/2024] Open
Abstract
Objective To develop a model combining clinical and radiomics features from CT scans for a preoperative noninvasive evaluation of Huvos grading of neoadjuvant chemotherapy in patients with HOS. Methods 183 patients from center A and 42 from center B were categorized into training and validation sets. Features derived from radiomics were obtained from unenhanced CT scans.Following dimensionality reduction, the most optimal features were selected and utilized in creating a radiomics model through logistic regression analysis. Integrating clinical features, a composite clinical radiomics model was developed, and a nomogram was constructed. Predictive performance of the model was evaluated using ROC curves and calibration curves. Additionally, decision curve analysis was conducted to assess practical utility of nomogram in clinical settings. Results LASSO LR analysis was performed, and finally, three selected image omics features were obtained.Radiomics model yielded AUC values with a good diagnostic effect for both patient sets (AUCs: 0.69 and 0.68, respectively). Clinical models (including sex, age, pre-chemotherapy ALP and LDH levels, new lung metastases within 1 year after surgery, and incidence) performed well in terms of Huvos grade prediction, with an AUC of 0.74 for training set. The AUC for independent validation set stood at 0.70. Notably, the amalgamation of radiomics and clinical features exhibited commendable predictive prowess in training set, registering an AUC of 0.78. This robust performance was subsequently validated in the independent validation set, where the AUC remained high at 0.75. Calibration curves of nomogram showed that the predictions were in good agreement with actual observations. Conclusion Combined model can be used for Huvos grading in patients with HOS after preoperative chemotherapy, which is helpful for adjuvant treatment decisions.
Collapse
Affiliation(s)
- Fan Yang
- Department of Radiation, Beijing Jishuitan Hospital,Capital Medical University, Beijing 100035, China
| | - Ying Feng
- Department of Ultrasound, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Pengfei Sun
- Department of Ultrasound, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Alberto Traverso
- Department of Radiation Oncology (Maastro), GROW-School for Oncology and Reproduction, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Andre Dekker
- Department of Radiation Oncology (Maastro), GROW-School for Oncology and Reproduction, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Bin Zhang
- Department of Radiation, Peking University Shougang Hospital, Beijing 100144, China
| | - Zhen Huang
- Department of Bone Oncology, Beijing Jishuitan Hospital,Capital Medical University, Beijing 100035, China
| | - Zhixiang Wang
- Department of Ultrasound, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Dong Yan
- Department of Radiation, Beijing Jishuitan Hospital,Capital Medical University, Beijing 100035, China
| |
Collapse
|
41
|
Trojani V, Bassi MC, Verzellesi L, Bertolini M. Impact of Preprocessing Parameters in Medical Imaging-Based Radiomic Studies: A Systematic Review. Cancers (Basel) 2024; 16:2668. [PMID: 39123396 PMCID: PMC11311340 DOI: 10.3390/cancers16152668] [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: 06/21/2024] [Revised: 07/16/2024] [Accepted: 07/24/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND Lately, radiomic studies featuring the development of a signature to use in prediction models in diagnosis or prognosis outcomes have been increasingly published. While the results are shown to be promising, these studies still have many pitfalls and limitations. One of the main issues of these studies is that radiomic features depend on how the images are preprocessed before their computation. Since, in widely known and used software for radiomic features calculation, it is possible to set these preprocessing parameters before the calculation of the radiomic feature, there are ongoing studies assessing the stability and repeatability of radiomic features to find the most suitable preprocessing parameters for every used imaging modality. MATERIALS AND METHODS We performed a comprehensive literature search using four electronic databases: PubMed, Cochrane Library, Embase, and Scopus. Mesh terms and free text were modeled in search strategies for databases. The inclusion criteria were studies where preprocessing parameters' influence on feature values and model predictions was addressed. Records lacking information on image acquisition parameters were excluded, and any eligible studies with full-text versions were included in the review process, while conference proceedings and monographs were disregarded. We used the QUADAS-2 (Quality Assessment of Diagnostic Accuracy Studies 2) tool to investigate the risk of bias. We synthesized our data in a table divided by the imaging modalities subgroups. RESULTS After applying the inclusion and exclusion criteria, we selected 43 works. This review examines the impact of preprocessing parameters on the reproducibility and reliability of radiomic features extracted from multimodality imaging (CT, MRI, CBCT, and PET/CT). Standardized preprocessing is crucial for consistent radiomic feature extraction. Key preprocessing steps include voxel resampling, normalization, and discretization, which influence feature robustness and reproducibility. In total, 44% of the included works studied the effects of an isotropic voxel resampling, and most studies opted to employ a discretization strategy. From 2021, several studies started selecting the best set of preprocessing parameters based on models' best performance. As for comparison metrics, ICC was the most used in MRI studies in 58% of the screened works. CONCLUSIONS From our work, we highlighted the need to harmonize the use of preprocessing parameters and their values, especially in light of future studies of prospective studies, which are still lacking in the current literature.
Collapse
Affiliation(s)
- Valeria Trojani
- Medical Physics, Azienda USL-IRCCS, 42123 Reggio Emilia, Italy; (L.V.); (M.B.)
| | | | - Laura Verzellesi
- Medical Physics, Azienda USL-IRCCS, 42123 Reggio Emilia, Italy; (L.V.); (M.B.)
| | - Marco Bertolini
- Medical Physics, Azienda USL-IRCCS, 42123 Reggio Emilia, Italy; (L.V.); (M.B.)
| |
Collapse
|
42
|
Ramlee S, Manavaki R, Aloj L, Escudero Sanchez L. Mitigating the impact of image processing variations on tumour [ 18F]-FDG-PET radiomic feature robustness. Sci Rep 2024; 14:16294. [PMID: 39009706 PMCID: PMC11251269 DOI: 10.1038/s41598-024-67239-8] [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: 01/30/2024] [Accepted: 07/09/2024] [Indexed: 07/17/2024] Open
Abstract
Radiomics analysis of [18F]-fluorodeoxyglucose ([18F]-FDG) PET images could be leveraged for personalised cancer medicine. However, the inherent sensitivity of radiomic features to intensity discretisation and voxel interpolation complicates its clinical translation. In this work, we evaluated the robustness of tumour [18F]-FDG-PET radiomic features to 174 different variations in intensity resolution or voxel size, and determined whether implementing parameter range conditions or dependency corrections could improve their robustness. Using 485 patient images spanning three cancer types: non-small cell lung cancer (NSCLC), melanoma, and lymphoma, we observed features were more sensitive to intensity discretisation than voxel interpolation, especially texture features. In most of our investigations, the majority of non-robust features could be made robust by applying parameter range conditions. Correctable features, which were generally fewer than conditionally robust, showed systematic dependence on bin configuration or voxel size that could be minimised by applying corrections based on simple mathematical equations. Melanoma images exhibited limited robustness and correctability relative to NSCLC and lymphoma. Our study provides an in-depth characterisation of the sensitivity of [18F]-FDG-PET features to image processing variations and reinforces the need for careful selection of imaging biomarkers prior to any clinical application.
Collapse
Affiliation(s)
- Syafiq Ramlee
- Department of Radiology, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK.
| | - Roido Manavaki
- Department of Radiology, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Luigi Aloj
- Department of Radiology, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- Department of Radiology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Lorena Escudero Sanchez
- Department of Radiology, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
| |
Collapse
|
43
|
Scicolone R, Vacca S, Pisu F, Benson JC, Nardi V, Lanzino G, Suri JS, Saba L. Radiomics and artificial intelligence: General notions and applications in the carotid vulnerable plaque. Eur J Radiol 2024; 176:111497. [PMID: 38749095 DOI: 10.1016/j.ejrad.2024.111497] [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: 03/16/2024] [Revised: 04/14/2024] [Accepted: 05/03/2024] [Indexed: 06/17/2024]
Abstract
Carotid atherosclerosis plays a substantial role in cardiovascular morbidity and mortality. Given the multifaceted impact of this disease, there has been increasing interest in harnessing artificial intelligence (AI) and radiomics as complementary tools for the quantitative analysis of medical imaging data. This integrated approach holds promise not only in refining medical imaging data analysis but also in optimizing the utilization of radiologists' expertise. By automating time consuming tasks, AI allows radiologists to focus on more pertinent responsibilities. Simultaneously, the capacity of AI in radiomics to extract nuanced patterns from raw data enhances the exploration of carotid atherosclerosis, advancing efforts in terms of (1) early detection and diagnosis, (2) risk stratification and predictive modeling, (3) improving workflow efficiency, and (4) contributing to advancements in research. This review provides an overview of general concepts related to radiomics and AI, along with their application in the field of carotid vulnerable plaque. It also offers insights into various research studies conducted on this topic across different imaging techniques.
Collapse
Affiliation(s)
- Roberta Scicolone
- Department of Radiology, Azienda Ospedaliero-Universitaria (A.O.U.), di Cagliari-Polo di Monserrato, Cagliari, Italy
| | - Sebastiano Vacca
- University of Cagliari, School of Medicine and Surgery, Cagliari, Italy
| | - Francesco Pisu
- Department of Radiology, Azienda Ospedaliero-Universitaria (A.O.U.), di Cagliari-Polo di Monserrato, Cagliari, Italy
| | - John C Benson
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Valentina Nardi
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Jasjit S Suri
- Stroke Monitoring and Diagnostic Division, AtheroPoint™, Roseville, CA, USA
| | - Luca Saba
- Department of Radiology, Azienda Ospedaliero-Universitaria (A.O.U.), di Cagliari-Polo di Monserrato, Cagliari, Italy.
| |
Collapse
|
44
|
Li X, Zhang L, Ding M. Ultrasound-based radiomics for the differential diagnosis of breast masses: A systematic review and meta-analysis. JOURNAL OF CLINICAL ULTRASOUND : JCU 2024; 52:778-788. [PMID: 38606802 DOI: 10.1002/jcu.23690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 02/19/2024] [Accepted: 04/01/2024] [Indexed: 04/13/2024]
Abstract
OBJECTIVES Ultrasound-based radiomics has demonstrated excellent diagnostic performance in differentiating benign and malignant breast masses. Given a few clinical studies on their diagnostic role, we conducted a meta-analysis of the potential effects of ultrasound-based radiomics for the differential diagnosis of breast masses, aiming to provide evidence-based medical basis for clinical research. MATERIALS AND METHODS We searched Embase, Web of Science, Cochrane Library, and PubMed databases from inception through to February 2023. The methodological quality assessment of the included studies was performed according to Quality Assessment of Diagnostic Accuracy Studies checklist. A diagnostic test accuracy systematic review and meta-analysis was performed in accordance with PRISMA guidelines. Sensitivity, specificity, and area under curve delineating benign and malignant lesions were recorded. We also used sensitivity analysis and subgroup analysis to explore potential sources of heterogeneity. Deeks' funnel plots was used to examine the publication bias. RESULTS A total of 11 studies were included in this meta-analysis. For the diagnosis of malignant breast masses worldwide, the overall mean rates of sensitivity and specificity of ultrasound-based radiomics were 0.90 (95% confidence interval [CI], 0.83-0.95) and 0.89 (95% CI, 0.82-0.94), respectively. The summary diagnostic odds ratio was 76 (95% CI, 26-219), and the area under the curve for the summary receiver operating characteristic curve was 0.95 (95% CI, 0.93-0.97). CONCLUSION Ultrasound-based radiomics has the potential to improve diagnostic accuracy to discriminate between benign and malignant breast masses, and could reduce unnecessary biopsies.
Collapse
Affiliation(s)
- Xuerong Li
- Hebei North University, Zhangjiakou, Hebei, China
| | | | - Manni Ding
- The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
45
|
Veiga-Canuto D, Fernández-Patón M, Cerdà Alberich L, Jiménez Pastor A, Gomis Maya A, Carot Sierra JM, Sangüesa Nebot C, Martínez de las Heras B, Pötschger U, Taschner-Mandl S, Neri E, Cañete A, Ladenstein R, Hero B, Alberich-Bayarri Á, Martí-Bonmatí L. Reproducibility Analysis of Radiomic Features on T2-weighted MR Images after Processing and Segmentation Alterations in Neuroblastoma Tumors. Radiol Artif Intell 2024; 6:e230208. [PMID: 38864742 PMCID: PMC11294951 DOI: 10.1148/ryai.230208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 04/22/2024] [Accepted: 05/30/2024] [Indexed: 06/13/2024]
Abstract
Purpose To evaluate the reproducibility of radiomics features extracted from T2-weighted MR images in patients with neuroblastoma. Materials and Methods A retrospective study included 419 patients (mean age, 29 months ± 34 [SD]; 220 male, 199 female) with neuroblastic tumors diagnosed between 2002 and 2023, within the scope of the PRedictive In-silico Multiscale Analytics to support cancer personalized diaGnosis and prognosis, Empowered by imaging biomarkers (ie, PRIMAGE) project, involving 746 T2/T2*-weighted MRI sequences at diagnosis and/or after initial chemotherapy. Images underwent processing steps (denoising, inhomogeneity bias field correction, normalization, and resampling). Tumors were automatically segmented, and 107 shape, first-order, and second-order radiomics features were extracted, considered as the reference standard. Subsequently, the previous image processing settings were modified, and volumetric masks were applied. New radiomics features were extracted and compared with the reference standard. Reproducibility was assessed using the concordance correlation coefficient (CCC); intrasubject repeatability was measured using the coefficient of variation (CoV). Results When normalization was omitted, only 5% of the radiomics features demonstrated high reproducibility. Statistical analysis revealed significant changes in the normalization and resampling processes (P < .001). Inhomogeneities removal had the least impact on radiomics (83% of parameters remained stable). Shape features remained stable after mask modifications, with a CCC greater than 0.90. Mask modifications were the most favorable changes for achieving high CCC values, with a radiomics features stability of 70%. Only 7% of second-order radiomics features showed an excellent CoV of less than 0.10. Conclusion Modifications in the T2-weighted MRI preparation process in patients with neuroblastoma resulted in changes in radiomics features, with normalization identified as the most influential factor for reproducibility. Inhomogeneities removal had the least impact on radiomics features. Keywords: Pediatrics, MR Imaging, Oncology, Radiomics, Reproducibility, Repeatability, Neuroblastic Tumors Supplemental material is available for this article. © RSNA, 2024 See also the commentary by Safdar and Galaria in this issue.
Collapse
Affiliation(s)
- Diana Veiga-Canuto
- From the Grupo de Investigación Biomédica en Imagen, Instituto de Investigación Sanitaria La Fe, Avenida Fernando Abril Martorell, 106 Torre A planta 7, 46026 Valencia, Spain (D.V.C., M.F.P., L.C.A., A.G.M., L.M.B.); Área Clínica de Imagen Médica (D.V.C., C.S.N., L.M.B.) and Department of Pediatric Oncology (B.M.d.l.H., A.C.), Hospital Universitari i Politècnic La Fe, Valencia, Spain; Quantitative Imaging Biomarkers in Medicine, QUIBIM SL, Valencia, Spain (A.J.P., A.A.B.); Departamento de Estadística e Investigación Operativa Aplicadas y Calidad, Universitat Politècnica de València, Valencia, Spain (J.M.C.S.); St. Anna Children’s Cancer Research Institute, Vienna, Austria (U.P., S.T.M., R.L.); Division of Academic Radiology, Department of Translational Research, University of Pisa, Pisa, Italy (E.N.); and Department of Pediatric Oncology and Hematology, University Children’s Hospital of Cologne, Medical 18 Faculty, University of Cologne, Cologne, Germany (B.H.)
| | - Matías Fernández-Patón
- From the Grupo de Investigación Biomédica en Imagen, Instituto de Investigación Sanitaria La Fe, Avenida Fernando Abril Martorell, 106 Torre A planta 7, 46026 Valencia, Spain (D.V.C., M.F.P., L.C.A., A.G.M., L.M.B.); Área Clínica de Imagen Médica (D.V.C., C.S.N., L.M.B.) and Department of Pediatric Oncology (B.M.d.l.H., A.C.), Hospital Universitari i Politècnic La Fe, Valencia, Spain; Quantitative Imaging Biomarkers in Medicine, QUIBIM SL, Valencia, Spain (A.J.P., A.A.B.); Departamento de Estadística e Investigación Operativa Aplicadas y Calidad, Universitat Politècnica de València, Valencia, Spain (J.M.C.S.); St. Anna Children’s Cancer Research Institute, Vienna, Austria (U.P., S.T.M., R.L.); Division of Academic Radiology, Department of Translational Research, University of Pisa, Pisa, Italy (E.N.); and Department of Pediatric Oncology and Hematology, University Children’s Hospital of Cologne, Medical 18 Faculty, University of Cologne, Cologne, Germany (B.H.)
| | - Leonor Cerdà Alberich
- From the Grupo de Investigación Biomédica en Imagen, Instituto de Investigación Sanitaria La Fe, Avenida Fernando Abril Martorell, 106 Torre A planta 7, 46026 Valencia, Spain (D.V.C., M.F.P., L.C.A., A.G.M., L.M.B.); Área Clínica de Imagen Médica (D.V.C., C.S.N., L.M.B.) and Department of Pediatric Oncology (B.M.d.l.H., A.C.), Hospital Universitari i Politècnic La Fe, Valencia, Spain; Quantitative Imaging Biomarkers in Medicine, QUIBIM SL, Valencia, Spain (A.J.P., A.A.B.); Departamento de Estadística e Investigación Operativa Aplicadas y Calidad, Universitat Politècnica de València, Valencia, Spain (J.M.C.S.); St. Anna Children’s Cancer Research Institute, Vienna, Austria (U.P., S.T.M., R.L.); Division of Academic Radiology, Department of Translational Research, University of Pisa, Pisa, Italy (E.N.); and Department of Pediatric Oncology and Hematology, University Children’s Hospital of Cologne, Medical 18 Faculty, University of Cologne, Cologne, Germany (B.H.)
| | - Ana Jiménez Pastor
- From the Grupo de Investigación Biomédica en Imagen, Instituto de Investigación Sanitaria La Fe, Avenida Fernando Abril Martorell, 106 Torre A planta 7, 46026 Valencia, Spain (D.V.C., M.F.P., L.C.A., A.G.M., L.M.B.); Área Clínica de Imagen Médica (D.V.C., C.S.N., L.M.B.) and Department of Pediatric Oncology (B.M.d.l.H., A.C.), Hospital Universitari i Politècnic La Fe, Valencia, Spain; Quantitative Imaging Biomarkers in Medicine, QUIBIM SL, Valencia, Spain (A.J.P., A.A.B.); Departamento de Estadística e Investigación Operativa Aplicadas y Calidad, Universitat Politècnica de València, Valencia, Spain (J.M.C.S.); St. Anna Children’s Cancer Research Institute, Vienna, Austria (U.P., S.T.M., R.L.); Division of Academic Radiology, Department of Translational Research, University of Pisa, Pisa, Italy (E.N.); and Department of Pediatric Oncology and Hematology, University Children’s Hospital of Cologne, Medical 18 Faculty, University of Cologne, Cologne, Germany (B.H.)
| | - Armando Gomis Maya
- From the Grupo de Investigación Biomédica en Imagen, Instituto de Investigación Sanitaria La Fe, Avenida Fernando Abril Martorell, 106 Torre A planta 7, 46026 Valencia, Spain (D.V.C., M.F.P., L.C.A., A.G.M., L.M.B.); Área Clínica de Imagen Médica (D.V.C., C.S.N., L.M.B.) and Department of Pediatric Oncology (B.M.d.l.H., A.C.), Hospital Universitari i Politècnic La Fe, Valencia, Spain; Quantitative Imaging Biomarkers in Medicine, QUIBIM SL, Valencia, Spain (A.J.P., A.A.B.); Departamento de Estadística e Investigación Operativa Aplicadas y Calidad, Universitat Politècnica de València, Valencia, Spain (J.M.C.S.); St. Anna Children’s Cancer Research Institute, Vienna, Austria (U.P., S.T.M., R.L.); Division of Academic Radiology, Department of Translational Research, University of Pisa, Pisa, Italy (E.N.); and Department of Pediatric Oncology and Hematology, University Children’s Hospital of Cologne, Medical 18 Faculty, University of Cologne, Cologne, Germany (B.H.)
| | - Jose Miguel Carot Sierra
- From the Grupo de Investigación Biomédica en Imagen, Instituto de Investigación Sanitaria La Fe, Avenida Fernando Abril Martorell, 106 Torre A planta 7, 46026 Valencia, Spain (D.V.C., M.F.P., L.C.A., A.G.M., L.M.B.); Área Clínica de Imagen Médica (D.V.C., C.S.N., L.M.B.) and Department of Pediatric Oncology (B.M.d.l.H., A.C.), Hospital Universitari i Politècnic La Fe, Valencia, Spain; Quantitative Imaging Biomarkers in Medicine, QUIBIM SL, Valencia, Spain (A.J.P., A.A.B.); Departamento de Estadística e Investigación Operativa Aplicadas y Calidad, Universitat Politècnica de València, Valencia, Spain (J.M.C.S.); St. Anna Children’s Cancer Research Institute, Vienna, Austria (U.P., S.T.M., R.L.); Division of Academic Radiology, Department of Translational Research, University of Pisa, Pisa, Italy (E.N.); and Department of Pediatric Oncology and Hematology, University Children’s Hospital of Cologne, Medical 18 Faculty, University of Cologne, Cologne, Germany (B.H.)
| | - Cinta Sangüesa Nebot
- From the Grupo de Investigación Biomédica en Imagen, Instituto de Investigación Sanitaria La Fe, Avenida Fernando Abril Martorell, 106 Torre A planta 7, 46026 Valencia, Spain (D.V.C., M.F.P., L.C.A., A.G.M., L.M.B.); Área Clínica de Imagen Médica (D.V.C., C.S.N., L.M.B.) and Department of Pediatric Oncology (B.M.d.l.H., A.C.), Hospital Universitari i Politècnic La Fe, Valencia, Spain; Quantitative Imaging Biomarkers in Medicine, QUIBIM SL, Valencia, Spain (A.J.P., A.A.B.); Departamento de Estadística e Investigación Operativa Aplicadas y Calidad, Universitat Politècnica de València, Valencia, Spain (J.M.C.S.); St. Anna Children’s Cancer Research Institute, Vienna, Austria (U.P., S.T.M., R.L.); Division of Academic Radiology, Department of Translational Research, University of Pisa, Pisa, Italy (E.N.); and Department of Pediatric Oncology and Hematology, University Children’s Hospital of Cologne, Medical 18 Faculty, University of Cologne, Cologne, Germany (B.H.)
| | - Blanca Martínez de las Heras
- From the Grupo de Investigación Biomédica en Imagen, Instituto de Investigación Sanitaria La Fe, Avenida Fernando Abril Martorell, 106 Torre A planta 7, 46026 Valencia, Spain (D.V.C., M.F.P., L.C.A., A.G.M., L.M.B.); Área Clínica de Imagen Médica (D.V.C., C.S.N., L.M.B.) and Department of Pediatric Oncology (B.M.d.l.H., A.C.), Hospital Universitari i Politècnic La Fe, Valencia, Spain; Quantitative Imaging Biomarkers in Medicine, QUIBIM SL, Valencia, Spain (A.J.P., A.A.B.); Departamento de Estadística e Investigación Operativa Aplicadas y Calidad, Universitat Politècnica de València, Valencia, Spain (J.M.C.S.); St. Anna Children’s Cancer Research Institute, Vienna, Austria (U.P., S.T.M., R.L.); Division of Academic Radiology, Department of Translational Research, University of Pisa, Pisa, Italy (E.N.); and Department of Pediatric Oncology and Hematology, University Children’s Hospital of Cologne, Medical 18 Faculty, University of Cologne, Cologne, Germany (B.H.)
| | - Ulrike Pötschger
- From the Grupo de Investigación Biomédica en Imagen, Instituto de Investigación Sanitaria La Fe, Avenida Fernando Abril Martorell, 106 Torre A planta 7, 46026 Valencia, Spain (D.V.C., M.F.P., L.C.A., A.G.M., L.M.B.); Área Clínica de Imagen Médica (D.V.C., C.S.N., L.M.B.) and Department of Pediatric Oncology (B.M.d.l.H., A.C.), Hospital Universitari i Politècnic La Fe, Valencia, Spain; Quantitative Imaging Biomarkers in Medicine, QUIBIM SL, Valencia, Spain (A.J.P., A.A.B.); Departamento de Estadística e Investigación Operativa Aplicadas y Calidad, Universitat Politècnica de València, Valencia, Spain (J.M.C.S.); St. Anna Children’s Cancer Research Institute, Vienna, Austria (U.P., S.T.M., R.L.); Division of Academic Radiology, Department of Translational Research, University of Pisa, Pisa, Italy (E.N.); and Department of Pediatric Oncology and Hematology, University Children’s Hospital of Cologne, Medical 18 Faculty, University of Cologne, Cologne, Germany (B.H.)
| | - Sabine Taschner-Mandl
- From the Grupo de Investigación Biomédica en Imagen, Instituto de Investigación Sanitaria La Fe, Avenida Fernando Abril Martorell, 106 Torre A planta 7, 46026 Valencia, Spain (D.V.C., M.F.P., L.C.A., A.G.M., L.M.B.); Área Clínica de Imagen Médica (D.V.C., C.S.N., L.M.B.) and Department of Pediatric Oncology (B.M.d.l.H., A.C.), Hospital Universitari i Politècnic La Fe, Valencia, Spain; Quantitative Imaging Biomarkers in Medicine, QUIBIM SL, Valencia, Spain (A.J.P., A.A.B.); Departamento de Estadística e Investigación Operativa Aplicadas y Calidad, Universitat Politècnica de València, Valencia, Spain (J.M.C.S.); St. Anna Children’s Cancer Research Institute, Vienna, Austria (U.P., S.T.M., R.L.); Division of Academic Radiology, Department of Translational Research, University of Pisa, Pisa, Italy (E.N.); and Department of Pediatric Oncology and Hematology, University Children’s Hospital of Cologne, Medical 18 Faculty, University of Cologne, Cologne, Germany (B.H.)
| | - Emanuele Neri
- From the Grupo de Investigación Biomédica en Imagen, Instituto de Investigación Sanitaria La Fe, Avenida Fernando Abril Martorell, 106 Torre A planta 7, 46026 Valencia, Spain (D.V.C., M.F.P., L.C.A., A.G.M., L.M.B.); Área Clínica de Imagen Médica (D.V.C., C.S.N., L.M.B.) and Department of Pediatric Oncology (B.M.d.l.H., A.C.), Hospital Universitari i Politècnic La Fe, Valencia, Spain; Quantitative Imaging Biomarkers in Medicine, QUIBIM SL, Valencia, Spain (A.J.P., A.A.B.); Departamento de Estadística e Investigación Operativa Aplicadas y Calidad, Universitat Politècnica de València, Valencia, Spain (J.M.C.S.); St. Anna Children’s Cancer Research Institute, Vienna, Austria (U.P., S.T.M., R.L.); Division of Academic Radiology, Department of Translational Research, University of Pisa, Pisa, Italy (E.N.); and Department of Pediatric Oncology and Hematology, University Children’s Hospital of Cologne, Medical 18 Faculty, University of Cologne, Cologne, Germany (B.H.)
| | - Adela Cañete
- From the Grupo de Investigación Biomédica en Imagen, Instituto de Investigación Sanitaria La Fe, Avenida Fernando Abril Martorell, 106 Torre A planta 7, 46026 Valencia, Spain (D.V.C., M.F.P., L.C.A., A.G.M., L.M.B.); Área Clínica de Imagen Médica (D.V.C., C.S.N., L.M.B.) and Department of Pediatric Oncology (B.M.d.l.H., A.C.), Hospital Universitari i Politècnic La Fe, Valencia, Spain; Quantitative Imaging Biomarkers in Medicine, QUIBIM SL, Valencia, Spain (A.J.P., A.A.B.); Departamento de Estadística e Investigación Operativa Aplicadas y Calidad, Universitat Politècnica de València, Valencia, Spain (J.M.C.S.); St. Anna Children’s Cancer Research Institute, Vienna, Austria (U.P., S.T.M., R.L.); Division of Academic Radiology, Department of Translational Research, University of Pisa, Pisa, Italy (E.N.); and Department of Pediatric Oncology and Hematology, University Children’s Hospital of Cologne, Medical 18 Faculty, University of Cologne, Cologne, Germany (B.H.)
| | - Ruth Ladenstein
- From the Grupo de Investigación Biomédica en Imagen, Instituto de Investigación Sanitaria La Fe, Avenida Fernando Abril Martorell, 106 Torre A planta 7, 46026 Valencia, Spain (D.V.C., M.F.P., L.C.A., A.G.M., L.M.B.); Área Clínica de Imagen Médica (D.V.C., C.S.N., L.M.B.) and Department of Pediatric Oncology (B.M.d.l.H., A.C.), Hospital Universitari i Politècnic La Fe, Valencia, Spain; Quantitative Imaging Biomarkers in Medicine, QUIBIM SL, Valencia, Spain (A.J.P., A.A.B.); Departamento de Estadística e Investigación Operativa Aplicadas y Calidad, Universitat Politècnica de València, Valencia, Spain (J.M.C.S.); St. Anna Children’s Cancer Research Institute, Vienna, Austria (U.P., S.T.M., R.L.); Division of Academic Radiology, Department of Translational Research, University of Pisa, Pisa, Italy (E.N.); and Department of Pediatric Oncology and Hematology, University Children’s Hospital of Cologne, Medical 18 Faculty, University of Cologne, Cologne, Germany (B.H.)
| | - Barbara Hero
- From the Grupo de Investigación Biomédica en Imagen, Instituto de Investigación Sanitaria La Fe, Avenida Fernando Abril Martorell, 106 Torre A planta 7, 46026 Valencia, Spain (D.V.C., M.F.P., L.C.A., A.G.M., L.M.B.); Área Clínica de Imagen Médica (D.V.C., C.S.N., L.M.B.) and Department of Pediatric Oncology (B.M.d.l.H., A.C.), Hospital Universitari i Politècnic La Fe, Valencia, Spain; Quantitative Imaging Biomarkers in Medicine, QUIBIM SL, Valencia, Spain (A.J.P., A.A.B.); Departamento de Estadística e Investigación Operativa Aplicadas y Calidad, Universitat Politècnica de València, Valencia, Spain (J.M.C.S.); St. Anna Children’s Cancer Research Institute, Vienna, Austria (U.P., S.T.M., R.L.); Division of Academic Radiology, Department of Translational Research, University of Pisa, Pisa, Italy (E.N.); and Department of Pediatric Oncology and Hematology, University Children’s Hospital of Cologne, Medical 18 Faculty, University of Cologne, Cologne, Germany (B.H.)
| | - Ángel Alberich-Bayarri
- From the Grupo de Investigación Biomédica en Imagen, Instituto de Investigación Sanitaria La Fe, Avenida Fernando Abril Martorell, 106 Torre A planta 7, 46026 Valencia, Spain (D.V.C., M.F.P., L.C.A., A.G.M., L.M.B.); Área Clínica de Imagen Médica (D.V.C., C.S.N., L.M.B.) and Department of Pediatric Oncology (B.M.d.l.H., A.C.), Hospital Universitari i Politècnic La Fe, Valencia, Spain; Quantitative Imaging Biomarkers in Medicine, QUIBIM SL, Valencia, Spain (A.J.P., A.A.B.); Departamento de Estadística e Investigación Operativa Aplicadas y Calidad, Universitat Politècnica de València, Valencia, Spain (J.M.C.S.); St. Anna Children’s Cancer Research Institute, Vienna, Austria (U.P., S.T.M., R.L.); Division of Academic Radiology, Department of Translational Research, University of Pisa, Pisa, Italy (E.N.); and Department of Pediatric Oncology and Hematology, University Children’s Hospital of Cologne, Medical 18 Faculty, University of Cologne, Cologne, Germany (B.H.)
| | - Luis Martí-Bonmatí
- From the Grupo de Investigación Biomédica en Imagen, Instituto de Investigación Sanitaria La Fe, Avenida Fernando Abril Martorell, 106 Torre A planta 7, 46026 Valencia, Spain (D.V.C., M.F.P., L.C.A., A.G.M., L.M.B.); Área Clínica de Imagen Médica (D.V.C., C.S.N., L.M.B.) and Department of Pediatric Oncology (B.M.d.l.H., A.C.), Hospital Universitari i Politècnic La Fe, Valencia, Spain; Quantitative Imaging Biomarkers in Medicine, QUIBIM SL, Valencia, Spain (A.J.P., A.A.B.); Departamento de Estadística e Investigación Operativa Aplicadas y Calidad, Universitat Politècnica de València, Valencia, Spain (J.M.C.S.); St. Anna Children’s Cancer Research Institute, Vienna, Austria (U.P., S.T.M., R.L.); Division of Academic Radiology, Department of Translational Research, University of Pisa, Pisa, Italy (E.N.); and Department of Pediatric Oncology and Hematology, University Children’s Hospital of Cologne, Medical 18 Faculty, University of Cologne, Cologne, Germany (B.H.)
| |
Collapse
|
46
|
Brown KH, Kerr BN, Pettigrew M, Connor K, Miller IS, Shiels L, Connolly C, McGarry C, Byrne AT, Butterworth KT. A comparative analysis of preclinical computed tomography radiomics using cone-beam and micro-computed tomography scanners. Phys Imaging Radiat Oncol 2024; 31:100615. [PMID: 39157293 PMCID: PMC11328005 DOI: 10.1016/j.phro.2024.100615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 07/17/2024] [Accepted: 07/20/2024] [Indexed: 08/20/2024] Open
Abstract
Background and purpose Radiomics analysis extracts quantitative data (features) from medical images. These features could potentially reflect biological characteristics and act as imaging biomarkers within precision medicine. However, there is a lack of cross-comparison and validation of radiomics outputs which is paramount for clinical implementation. In this study, we compared radiomics outputs across two computed tomography (CT)-based preclinical scanners. Materials and methods Cone beam CT (CBCT) and µCT scans were acquired using different preclinical CT imaging platforms. The reproducibility of radiomics features on each scanner was assessed using a phantom across imaging energies (40 & 60 kVp) and segmentation volumes (44-238 mm3). Retrospective mouse scans were used to compare feature reliability across varying tissue densities (lung, heart, bone), scanners and after voxel size harmonisation. Reliable features had an intraclass correlation coefficient (ICC) > 0.8. Results First order and GLCM features were the most reliable on both scanners across different volumes. There was an inverse relationship between tissue density and feature reliability, with the highest number of features in lung (CBCT=580, µCT=734) and lowest in bone (CBCT=110, µCT=560). Comparable features for lung and heart tissues increased when voxel sizes were harmonised. We have identified tissue-specific preclinical radiomics signatures in mice for the lung (133), heart (35), and bone (15). Conclusions Preclinical CBCT and µCT scans can be used for radiomics analysis to support the development of meaningful radiomics signatures. This study demonstrates the importance of standardisation and emphasises the need for multi-centre studies.
Collapse
Affiliation(s)
- Kathryn H Brown
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, United Kingdom
| | - Brianna N Kerr
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, United Kingdom
| | - Mihaela Pettigrew
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, United Kingdom
| | - Kate Connor
- Department of Physiology and Medical Physics and Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Ian S Miller
- Department of Physiology and Medical Physics and Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin 2, Ireland
- National Preclinical Imaging Centre, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Liam Shiels
- Department of Physiology and Medical Physics and Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Colum Connolly
- Department of Physiology and Medical Physics and Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Conor McGarry
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, United Kingdom
- Northern Ireland Cancer Centre, Belfast Health & Social Care Trust, Belfast, United Kingdom
| | - Annette T Byrne
- Department of Physiology and Medical Physics and Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin 2, Ireland
- National Preclinical Imaging Centre, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Karl T Butterworth
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, United Kingdom
| |
Collapse
|
47
|
Bortolotto C, Pinto A, Brero F, Messana G, Cabini RF, Postuma I, Robustelli Test A, Stella GM, Galli G, Mariani M, Figini S, Lascialfari A, Filippi AR, Bottinelli OM, Preda L. CT and MRI radiomic features of lung cancer (NSCLC): comparison and software consistency. Eur Radiol Exp 2024; 8:71. [PMID: 38880866 PMCID: PMC11180643 DOI: 10.1186/s41747-024-00468-8] [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: 01/31/2024] [Accepted: 04/10/2024] [Indexed: 06/18/2024] Open
Abstract
BACKGROUND Radiomics is a quantitative approach that allows the extraction of mineable data from medical images. Despite the growing clinical interest, radiomics studies are affected by variability stemming from analysis choices. We aimed to investigate the agreement between two open-source radiomics software for both contrast-enhanced computed tomography (CT) and contrast-enhanced magnetic resonance imaging (MRI) of lung cancers and to preliminarily evaluate the existence of radiomic features stable for both techniques. METHODS Contrast-enhanced CT and MRI images of 35 patients affected with non-small cell lung cancer (NSCLC) were manually segmented and preprocessed using three different methods. Sixty-six Image Biomarker Standardisation Initiative-compliant features common to the considered platforms, PyRadiomics and LIFEx, were extracted. The correlation among features with the same mathematical definition was analyzed by comparing PyRadiomics and LIFEx (at fixed imaging technique), and MRI with CT results (for the same software). RESULTS When assessing the agreement between LIFEx and PyRadiomics across the considered resampling, the maximum statistically significant correlations were observed to be 94% for CT features and 95% for MRI ones. When examining the correlation between features extracted from contrast-enhanced CT and MRI using the same software, higher significant correspondences were identified in 11% of features for both software. CONCLUSIONS Considering NSCLC, (i) for both imaging techniques, LIFEx and PyRadiomics agreed on average for 90% of features, with MRI being more affected by resampling and (ii) CT and MRI contained mostly non-redundant information, but there are shape features and, more importantly, texture features that can be singled out by both techniques. RELEVANCE STATEMENT Identifying and selecting features that are stable cross-modalities may be one of the strategies to pave the way for radiomics clinical translation. KEY POINTS • More than 90% of LIFEx and PyRadiomics features contain the same information. • Ten percent of features (shape, texture) are stable among contrast-enhanced CT and MRI. • Software compliance and cross-modalities stability features are impacted by the resampling method.
Collapse
Affiliation(s)
- Chandra Bortolotto
- Radiology Institute, Fondazione IRCCS Policlinico San Matteo, Pavia, 27100, Italy
- Diagnostic Imaging and Radiotherapy Unit, Department of Clinical, Surgical, Diagnostic, and Pediatric Sciences, University of Pavia, Pavia, 27100, Italy
| | - Alessandra Pinto
- Radiology Institute, Fondazione IRCCS Policlinico San Matteo, Pavia, 27100, Italy.
| | - Francesca Brero
- Department of Physics, University of Pavia, Via Bassi 6, Pavia, 27100, Italy
- Istituto Nazionale Di Fisica Nucleare, Sezione Di Pavia, Pavia, 27100, Italy
| | - Gaia Messana
- Radiology Institute, Fondazione IRCCS Policlinico San Matteo, Pavia, 27100, Italy
| | - Raffaella Fiamma Cabini
- Istituto Nazionale Di Fisica Nucleare, Sezione Di Pavia, Pavia, 27100, Italy.
- Department of Mathematics, University of Pavia, Via Ferrata 5, Pavia, 27100, Italy.
| | - Ian Postuma
- Istituto Nazionale Di Fisica Nucleare, Sezione Di Pavia, Pavia, 27100, Italy
| | - Agnese Robustelli Test
- Department of Physics, University of Pavia, Via Bassi 6, Pavia, 27100, Italy.
- Istituto Nazionale Di Fisica Nucleare, Sezione Di Pavia, Pavia, 27100, Italy.
| | - Giulia Maria Stella
- Department of Medical Sciences and Infective Diseases, Unit of Respiratory Diseases, Fondazione IRCCS Policlinico San Matteo, Pavia, 27100, Italy
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, 27100, Italy
| | - Giulia Galli
- Department of Medical Sciences and Infective Diseases, Unit of Respiratory Diseases, Fondazione IRCCS Policlinico San Matteo, Pavia, 27100, Italy
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, 27100, Italy
| | - Manuel Mariani
- Department of Physics, University of Pavia, Via Bassi 6, Pavia, 27100, Italy
| | - Silvia Figini
- Department of Political and Social Sciences, University of Pavia, Pavia, 27100, Italy
| | - Alessandro Lascialfari
- Department of Physics, University of Pavia, Via Bassi 6, Pavia, 27100, Italy
- Istituto Nazionale Di Fisica Nucleare, Sezione Di Pavia, Pavia, 27100, Italy
| | - Andrea Riccardo Filippi
- Diagnostic Imaging and Radiotherapy Unit, Department of Clinical, Surgical, Diagnostic, and Pediatric Sciences, University of Pavia, Pavia, 27100, Italy
- Department of Radiation Oncology, Fondazione IRCCS Policlinico San Matteo, Pavia, 27100, Italy
| | - Olivia Maria Bottinelli
- Diagnostic Imaging and Radiotherapy Unit, Department of Clinical, Surgical, Diagnostic, and Pediatric Sciences, University of Pavia, Pavia, 27100, Italy
| | - Lorenzo Preda
- Radiology Institute, Fondazione IRCCS Policlinico San Matteo, Pavia, 27100, Italy
- Diagnostic Imaging and Radiotherapy Unit, Department of Clinical, Surgical, Diagnostic, and Pediatric Sciences, University of Pavia, Pavia, 27100, Italy
| |
Collapse
|
48
|
Zhang X, Iqbal Bin Saripan M, Wu Y, Wang Z, Wen D, Cao Z, Wang B, Xu S, Liu Y, Marhaban MH, Dong X. The impact of the combat method on radiomics feature compensation and analysis of scanners from different manufacturers. BMC Med Imaging 2024; 24:137. [PMID: 38844854 PMCID: PMC11157873 DOI: 10.1186/s12880-024-01306-4] [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: 01/02/2023] [Accepted: 05/23/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND This study investigated whether the Combat compensation method can remove the variability of radiomic features extracted from different scanners, while also examining its impact on the subsequent predictive performance of machine learning models. MATERIALS AND METHODS 135 CT images of Credence Cartridge Radiomic phantoms were collected and screened from three scanners manufactured by Siemens, Philips, and GE. 100 radiomic features were extracted and 20 radiomic features were screened according to the Lasso regression method. The radiomic features extracted from the rubber and resin-filled regions in the cartridges were labeled into different categories for evaluating the performance of the machine learning model. Radiomics features were divided into three groups based on the different scanner manufacturers. The radiomic features were randomly divided into training and test sets with a ratio of 8:2. Five machine learning models (lasso, logistic regression, random forest, support vector machine, neural network) were employed to evaluate the impact of Combat on radiomic features. The variability among radiomic features were assessed using analysis of variance (ANOVA) and principal component analysis (PCA). Accuracy, precision, recall, and area under the receiver curve (AUC) were used as evaluation metrics for model classification. RESULTS The principal component and ANOVA analysis results show that the variability of different scanner manufacturers in radiomic features was removed (P˃0.05). After harmonization with the Combat algorithm, the distributions of radiomic features were aligned in terms of location and scale. The performance of machine learning models for classification improved, with the Random Forest model showing the most significant enhancement. The AUC value increased from 0.88 to 0.92. CONCLUSIONS The Combat algorithm has reduced variability in radiomic features from different scanners. In the phantom CT dataset, it appears that the machine learning model's classification performance may have improved after Combat harmonization. However, further investigation and validation are required to fully comprehend Combat's impact on radiomic features in medical imaging.
Collapse
Affiliation(s)
- Xiaolei Zhang
- Faculty of Engineering, Universiti Putra Malaysia, Serdang, Malaysia.
- Hebei International Research Center of Medical Engineering, Chengde Medical University, Chengde City, Hebei Province, China.
- Department of Biomedical Engineering, Chengde Medical University, Chengde City, Hebei Province, China.
| | | | - Yanjun Wu
- Hebei International Research Center of Medical Engineering, Chengde Medical University, Chengde City, Hebei Province, China
| | - Zhongxiao Wang
- Hebei International Research Center of Medical Engineering, Chengde Medical University, Chengde City, Hebei Province, China
| | - Dong Wen
- Institute of Artificial Intelligence, University of Science and Technology Beijing, Beijing, China
| | - Zhendong Cao
- Department of Radiology, the Affiliated Hospital of Chengde Medical University, Chengde, Hebei, China
| | - Bingzhen Wang
- Faculty of Engineering, Universiti Putra Malaysia, Serdang, Malaysia
- Hebei International Research Center of Medical Engineering, Chengde Medical University, Chengde City, Hebei Province, China
| | - Shiqi Xu
- Hebei International Research Center of Medical Engineering, Chengde Medical University, Chengde City, Hebei Province, China
| | - Yanli Liu
- Hebei International Research Center of Medical Engineering, Chengde Medical University, Chengde City, Hebei Province, China
| | | | - Xianling Dong
- Hebei International Research Center of Medical Engineering, Chengde Medical University, Chengde City, Hebei Province, China.
- Hebei Provincial Key Laboratory of Nerve Injury and Repair, Chengde Medical University, Chengde City, Hebei Province, China.
| |
Collapse
|
49
|
Zhang Y, Yang X, Bi F, Wen L, Niu Y, Yang Y, Lin H, Yu X. CT-based radiomics for differentiating peripherally located pulmonary sclerosing pneumocytoma from carcinoid. Med Phys 2024; 51:4219-4230. [PMID: 38507783 DOI: 10.1002/mp.17037] [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: 09/05/2023] [Revised: 01/31/2024] [Accepted: 03/07/2024] [Indexed: 03/22/2024] Open
Abstract
BACKGROUND Pulmonary sclerosing pneumocytoma (PSP) and pulmonary carcinoid (PC) are difficult to distinguish based on conventional imaging examinations. In recent years, radiomics has been used to discriminate benign from malignant pulmonary lesions. However, the value of radiomics based on computed tomography (CT) images to differentiate PSP from PC has not been well explored. PURPOSE We aimed to investigate the feasibility of radiomics in the differentiation between PSP and PC. METHODS Fifty-three PSP and fifty-five PC were retrospectively enrolled and then were randomly divided into the training and test sets. Univariate and multivariable logistic analyses were carried to select clinical predictor related to differential diagnosis of PSP and PC. A total of 1316 radiomics features were extracted from the unenhanced CT (UECT) and contrast-enhanced CT (CECT) images, respectively. The minimum redundancy maximum relevance and the least absolute shrinkage and selection operator were used to select the most significant radiomics features to construct radiomics models. The clinical predictor and radiomics features were integrated to develop combined models. Two senior radiologists independently categorized each patient into PSP or PC group based on traditional CT method. The performances of clinical, radiomics, and combined models in differentiating PSP from PC were investigated by the receiver operating characteristic (ROC) curve. The diagnostic performance was also compared between the combined models and radiologists. RESULTS In regard to differentiating PSP from PC, the area under the curves (AUCs) of the clinical, radiomics, and combined models were 0.87, 0.96, and 0.99 in the training set UECT, and were 0.87, 0.97, and 0.98 in the training set CECT, respectively. The AUCs of the clinical, radiomics, and combined models were 0.84, 0.92, and 0.97 in the test set UECT, and were 0.84, 0.93, and 0.98 in the test set CECT, respectively. In regard to the differentiation between PSP and PC, the combined model was comparable to the radiomics model, but outperformed the clinical model and the two radiologists, whether in the test set UECT or CECT. CONCLUSIONS Radiomics approaches show promise in distinguishing between PSP and PC. Moreover, the integration of clinical predictor (gender) has the potential to enhance the diagnostic performance even further.
Collapse
Affiliation(s)
- Yi Zhang
- Graduate Collaborative Training base of Hunan Cancer Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Department of Diagnostic Radiology, The Affiliated Cancer Hospital of Xiangya School of Medicine & Hunan Cancer Hospital, Central South University, Changsha, Hunan, China
| | - Xiaohuang Yang
- Department of Diagnostic Radiology, The Affiliated Cancer Hospital of Xiangya School of Medicine & Hunan Cancer Hospital, Central South University, Changsha, Hunan, China
| | - Feng Bi
- Department of Diagnostic Radiology, The Affiliated Cancer Hospital of Xiangya School of Medicine & Hunan Cancer Hospital, Central South University, Changsha, Hunan, China
| | - Lu Wen
- Department of Diagnostic Radiology, The Affiliated Cancer Hospital of Xiangya School of Medicine & Hunan Cancer Hospital, Central South University, Changsha, Hunan, China
| | - Yue Niu
- Graduate Collaborative Training base of Hunan Cancer Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yanhui Yang
- Graduate Collaborative Training base of Hunan Cancer Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Huashan Lin
- Department of Pharmaceutical Diagnosis, General Electric (GE) Healthcare, Changsha, Hunan, China
| | - Xiaoping Yu
- Graduate Collaborative Training base of Hunan Cancer Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Department of Diagnostic Radiology, The Affiliated Cancer Hospital of Xiangya School of Medicine & Hunan Cancer Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
50
|
Li M, Yuan Y, Zhou H, Feng F, Xu G. A multicenter study: predicting KRAS mutation and prognosis in colorectal cancer through a CT-based radiomics nomogram. Abdom Radiol (NY) 2024; 49:1816-1828. [PMID: 38393357 DOI: 10.1007/s00261-024-04218-7] [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: 11/23/2023] [Revised: 01/16/2024] [Accepted: 01/17/2024] [Indexed: 02/25/2024]
Abstract
PURPOSE To establish a CT-based radiomics nomogram for preoperative prediction of KRAS mutation and prognostic stratification in colorectal cancer (CRC) patients. METHODS In a retrospective analysis, 408 patients with confirmed CRC were included, comprising 168 cases in the training set, 111 cases in the internal validation set, and 129 cases in the external validation set. Radiomics features extracted from the primary tumors were meticulously screened to identify those closely associated with KRAS mutation. Subsequently, a radiomics nomogram was constructed by integrating these radiomics features with clinically significant parameters. The diagnostic performance was assessed through the area under the receiver operating characteristic curve (AUC). Lastly, the prognostic significance of the nomogram was explored, and Kaplan-Meier analysis was employed to depict survival curves for the high-risk and low-risk groups. RESULTS A radiomics model was constructed using 19 radiomics features significantly associated with KRAS mutation. Furthermore, a nomogram was developed by integrating these radiomics features with two clinically significant parameters (age, tumor location). The nomogram achieved AUCs of 0.834, 0.813, and 0.811 in the training set, internal validation set, and external validation set, respectively. Additionally, the nomogram effectively stratified patients into high-risk (KRAS mutation) and low-risk (KRAS wild-type) groups, demonstrating a significant difference in overall survival (P < 0.001). Patients categorized in the high-risk group exhibited inferior overall survival in contrast to those classified in the low-risk group. CONCLUSIONS The CT-based radiomics nomogram demonstrates the capability to effectively predict KRAS mutation in CRC patients and stratify their prognosis preoperatively.
Collapse
Affiliation(s)
- Manman Li
- Department of Radiology, Yancheng No 1 People's Hospital, The Fourth Affiliated Hospital of Nantong University, Yancheng, Jiangsu Province, 224006, China
| | - Yiwen Yuan
- Department of Translational Medical Center, Yancheng No 1 People's Hospital, The Fourth Affiliated Hospital of Nantong University, Yancheng, Jiangsu Province, 224006, China
| | - Hui Zhou
- Department of Radiology, Affiliated Tumor Hospital of Nantong University, Nantong, Jiangsu Province, 226001, China
| | - Feng Feng
- Department of Radiology, Affiliated Tumor Hospital of Nantong University, Nantong, Jiangsu Province, 226001, China.
| | - Guodong Xu
- Department of Radiology, Yancheng No 1 People's Hospital, The Fourth Affiliated Hospital of Nantong University, Yancheng, Jiangsu Province, 224006, China.
| |
Collapse
|